Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Neuroimmune Pharmacol ; 19(1): 29, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874861

ABSTRACT

The opioid epidemic has received considerable attention, but the impact on perinatal opioid-exposed (POE) offspring remains underexplored. This study addresses the emerging public health challenge of understanding and treating POE children. We examined two scenarios using preclinical models: offspring exposed to oxycodone (OXY) in utero (IUO) and acute postnatal OXY (PNO). We hypothesized exposure to OXY during pregnancy primes offspring for neurodevelopmental deficits and severity of deficits is dependent on timing of exposure. Notable findings include reduced head size and brain weight in offspring. Molecular analyses revealed significantly lower levels of inflammasome-specific genes in the prefrontal cortex (PFC). Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) highlighted the enrichment of genes associated with mitochondrial and synapse dysfunction in POE offspring. Western blot analysis validated IPA predictions of mitochondrial dysfunction in PFC-derived synaptosomes. Behavioral studies identified significant social deficits in POE offspring. This study presents the first comparative analysis of acute PNO- and IUO-offspring during early adolescence finding acute PNO-offspring have considerably greater deficits. The striking difference in deficit severity in acute PNO-offspring suggests that exposure to opioids in late pregnancy pose the greatest risk for offspring well-being.


Subject(s)
Analgesics, Opioid , Oxycodone , Prenatal Exposure Delayed Effects , Animals , Oxycodone/toxicity , Pregnancy , Female , Prenatal Exposure Delayed Effects/chemically induced , Male , Analgesics, Opioid/adverse effects , Analgesics, Opioid/toxicity , Behavior, Animal/drug effects , Rats , Rats, Sprague-Dawley , Neurodevelopmental Disorders/chemically induced , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
2.
J Pharmacol Exp Ther ; 380(1): 1-14, 2022 01.
Article in English | MEDLINE | ID: mdl-34625464

ABSTRACT

Opioid use disorder reflects a major public health crisis of morbidity and mortality in which opioid withdrawal often contributes to continued use. However, current medications that treat opioid withdrawal symptoms are limited by their abuse liability or lack of efficacy. Although cannabinoid 1 (CB1) receptor agonists, including Δ9-tetrahydrocannabinol, ameliorate opioid withdrawal in both clinical and preclinical studies of opioid dependence, this strategy elicits cannabimimetic side effects as well as tolerance and dependence after repeated administration. Alternatively, CB1 receptor positive allosteric modulators (PAMs) enhance CB1 receptor signaling and show efficacy in rodent models of pain and cannabinoid dependence but lack cannabimimetic side effects. We hypothesize that the CB1 receptor PAM ZCZ011 attenuates naloxone-precipitated withdrawal signs in opioid-dependent mice. Accordingly, male and female mice given an escalating dosing regimen of oxycodone, a widely prescribed opioid, and challenged with naloxone displayed withdrawal signs that included diarrhea, weight loss, jumping, paw flutters, and head shakes. ZCZ011 fully attenuated naloxone-precipitated withdrawal-induced diarrhea and weight loss and reduced paw flutters by approximately half, but its effects on head shakes were unreliable, and it did not affect jumping behavior. The antidiarrheal and anti-weight loss effects of ZCZ0111 were reversed by a CB1 not a cannabinoid receptor type 2 receptor antagonist and were absent in CB1 (-/-) mice, suggesting a necessary role of CB1 receptors. Collectively, these results indicate that ZCZ011 completely blocked naloxone-precipitated diarrhea and weight loss in oxycodone-dependent mice and suggest that CB1 receptor PAMs may offer a novel strategy to treat opioid dependence. SIGNIFICANCE STATEMENT: Opioid use disorder represents a serious public health crisis in which current medications used to treat withdrawal symptoms are limited by abuse liability and side effects. The CB1 receptor positive allosteric modulator (PAM) ZCZ011, which lacks overt cannabimimetic behavioral effects, ameliorated naloxone-precipitated withdrawal signs through a CB1 receptor mechanism of action in a mouse model of oxycodone dependence. These results suggest that CB1 receptor PAMs may represent a viable strategy to treat opioid withdrawal.


Subject(s)
Antidiarrheals/therapeutic use , Cannabinoid Receptor Agonists/therapeutic use , Diarrhea/drug therapy , Indoles/therapeutic use , Substance Withdrawal Syndrome/drug therapy , Thiophenes/therapeutic use , Allosteric Regulation , Animals , Diarrhea/etiology , Female , Male , Mice , Mice, Inbred ICR , Naloxone/adverse effects , Narcotic Antagonists/adverse effects , Narcotics/toxicity , Opioid-Related Disorders/drug therapy , Opioid-Related Disorders/etiology , Oxycodone/toxicity , Receptor, Cannabinoid, CB1/metabolism , Substance Withdrawal Syndrome/etiology
3.
Genes Brain Behav ; 20(8): e12770, 2021 11.
Article in English | MEDLINE | ID: mdl-34459088

ABSTRACT

The steady rise in prescription opioids such as oxycodone has led to a virulent epidemic of widespread abuse and deaths in the United States; approximately 80% of affected individuals initiate the habitual use of oxycodone by using prescription oral oxycodone. Given the importance of drug pharmacokinetics in determining abuse potential, we designed an oral operant oxycodone self-administration (SA) procedure in rats to model drug intake by most human users/abusers of oxycodone. Key aspects of the model include limited initial drug intake followed by increasing drug concentrations during extended 4-h sessions on alternating days. Sex and genetic predisposition are major determinants of human opiate abuse. Therefore, we studied females in seven inbred strains (WLI, WMI, LEW, DSS, F344, BN and SHR) and both sexes in three of these strains. All strains increased intake across serially increasing doses (0.025-0.2 mg/ml; p < 0.001): the range of intakes at the final concentration of oxycodone was 0.72 ± 0.17-4.84 ± 1.42 mg/kg (mean ± SEM) - a 6.7-fold difference across strains. In LEW, WLI and WMI strains, oxycodone intake increased significantly across all sessions in both sexes. However, in LEW and WMI male rats but not WLI, daily oxycodone intake was significantly lower across all 4-h sessions than females (p < 0.005). The estimated heritability in oxycodone intake was in the range of 0.21-0.41. In summary, our novel operant oral oxycodone SA model captures the strong abuse potential of oral oxycodone and shows dose, sex and strain-specific drug intake that is significantly dependent on heredity.


Subject(s)
Analgesics, Opioid/administration & dosage , Disease Models, Animal , Genetic Predisposition to Disease , Opioid-Related Disorders/genetics , Oxycodone/administration & dosage , Analgesics, Opioid/toxicity , Animals , Female , Male , Opioid-Related Disorders/physiopathology , Oxycodone/toxicity , Rats , Rats, Inbred Dahl , Rats, Inbred F344 , Rats, Inbred Lew , Rats, Wistar , Self Administration , Sex
4.
J Pharmacol Exp Ther ; 377(2): 232-241, 2021 05.
Article in English | MEDLINE | ID: mdl-33622770

ABSTRACT

This study evaluated a battery of pain-stimulated, pain-depressed, and pain-independent behaviors for preclinical pharmacological assessment of candidate analgesics in mice. Intraperitoneal injection of dilute lactic acid (IP acid) served as an acute visceral noxious stimulus to produce four pain-related behaviors in male and female ICR mice: stimulation of 1) stretching, 2) facial grimace, 3) depression of rearing, and 4) depression of nesting. Additionally, nesting and locomotion in the absence of the noxious stimulus were used to assess pain-independent drug effects. These six behaviors were used to compare effects of two mechanistically distinct but clinically effective positive controls (ketoprofen and oxycodone) and two negative controls that are not clinically approved as analgesics but produce either general motor depression (diazepam) or motor stimulation (amphetamine). We predicted that analgesics would alleviate all IP acid effects at doses that did not alter pain-independent behaviors, whereas negative controls would not. Consistent with this prediction, ketoprofen (0.1-32 mg/kg) produced the expected analgesic profile, whereas oxycodone (0.32-3.2 mg/kg) alleviated all IP acid effects except depression of rearing at doses lower than those that altered pain-independent behaviors. For the negative controls, diazepam (1-10 mg/kg) failed to block IP acid-induced depression of either rearing or nesting and only decreased IP acid-stimulated behaviors at doses that also decreased pain-independent behaviors. Amphetamine (0.32-3.2 mg/kg) alleviated all IP acid effects but only at doses that also stimulated locomotion. These results support utility of this model as a framework to evaluate candidate-analgesic effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behavioral endpoints. SIGNIFICANCE STATEMENT: Preclinical assays of pain and analgesia often yield false-positive effects with candidate analgesics. This study used two positive-control analgesics (ketoprofen, oxycodone) and two active negative controls (diazepam, amphetamine) to validate a strategy for distinguishing analgesics from nonanalgesics by profiling drug effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behaviors in male and female mice.


Subject(s)
Analgesics/toxicity , Behavior, Animal , Movement , Pain/drug therapy , Amphetamine/administration & dosage , Amphetamine/therapeutic use , Amphetamine/toxicity , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Diazepam/administration & dosage , Diazepam/therapeutic use , Diazepam/toxicity , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , False Negative Reactions , Female , Ketoprofen/administration & dosage , Ketoprofen/therapeutic use , Ketoprofen/toxicity , Male , Mice , Mice, Inbred ICR , No-Observed-Adverse-Effect Level , Oxycodone/administration & dosage , Oxycodone/therapeutic use , Oxycodone/toxicity
5.
Neuropharmacology ; 186: 108469, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33485944

ABSTRACT

Opioid use disorder is a leading cause of morbidity and mortality in the United States. Increasing pre-clinical and clinical evidence demonstrates sex differences in opioid use and dependence. However, the underlying molecular mechanisms contributing to these effects, including neuroinflammation, are still obscure. Therefore, in this study, we investigated the effect of oxycodone exposure and withdrawal on sex- and region-specific neuroimmune response. Real-time PCR and multiplex cytokine array analysis demonstrated elevated neuroinflammation with increased pro-inflammatory cytokine levels, and aberrant oligodendroglial response in reward neurocircuitry, following withdrawal from chronic oxycodone treatment. Chronic oxycodone and withdrawal treated male mice had lower mRNA expression of TMEM119 along with elevated protein levels of pro-inflammatory cytokines/chemokines and growth factors (IL-1ß, IL-2, IL-7, IL-9, IL-12, IL-15, IL17, M-CSF, VEGF) in the prefrontal cortex (PFC) as compared to their female counterparts. In contrast, reduced levels of pro-inflammatory cytokines/chemokines (IL-1ß, IL-6, IL-9, IL-12, CCL11) was observed in the nucleus accumbens (NAc) of oxycodone and withdrawal-treated males as compared to female mice. No treatment specific effects were observed on the mRNA expression of putative microglial activation markers (Iba1, CD68), but an overall sex specific decrease in the mRNA expression of Iba1 and CD68 was found in the PFC and NAc of male mice as compared to females. Moreover, a sex and region-specific increase in the mRNA levels of oligodendrocyte lineage markers (NG2, Sox10) was also observed in oxycodone and withdrawal treated animals. These findings may open a new avenue for the development of sex-specific precision therapeutics for opioid dependence by targeting region-specific neuroimmune signaling.


Subject(s)
Analgesics, Opioid/administration & dosage , Analgesics, Opioid/toxicity , Neuroimmunomodulation/immunology , Sex Characteristics , Substance Withdrawal Syndrome/immunology , Substance Withdrawal Syndrome/metabolism , Animals , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neuroimmunomodulation/drug effects , Oxycodone/administration & dosage , Oxycodone/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/immunology , Prefrontal Cortex/metabolism , Substance Withdrawal Syndrome/diagnosis
6.
Biosci Rep ; 40(2)2020 02 28.
Article in English | MEDLINE | ID: mdl-31967294

ABSTRACT

BACKGROUND: Oxycodone is an opioid medication used for the treatment of pain in cancer patients. However, little is known on the direct effects of oxycodone on cancer cells. AIM: To determine the effects and mechanisms of oxycodone in cancer cells. MATERIALS AND METHODS: Proliferation, survival and migration assays were performed on multiple types of cancer cells. Epithelial growth factor receptor (EGFR)/ERK/Akt pathway and oxidative stress were investigated after oxycodone treatment. RESULTS: Oxycodone can either stimulate growth and migration without affecting survival in MDA-468 cells or inhibit growth and survival without affecting migration in SKBR3 and Caco2 cells. In addition, oxycodone can either attenuate or stimulate efficacy of chemotherapeutic drugs in cancer, depending on the type of cancer cells and nature of action of oxycodone as single drug alone. Our mechanism studies suggest that the stimulatory and inhibitory effects of oxycodone are associated with EGFR expression levels in cancer cells. In cancer cells with high EGFR level, oxycodone activates EGFR signaling in cancer cells, leading to stimulatory effects in multiple biological activities, and this is dependent on opioid receptor. In cancer cells with low EGFR level, oxycodone induces mitochondria-mediated caspase activity and oxidative stress and damage, leading to cell death. CONCLUSIONS: Our work is the first to demonstrate systematic analysis of oxycodone's effects and mechanism of action in cancer. The activation of EGFR signaling by oxycodone may provide a new guide in the clinical use of oxycodone, in particular for cancer patients with high EGFR levels.


Subject(s)
Analgesics, Opioid/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/drug therapy , Oxycodone/pharmacology , Analgesics, Opioid/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Apoptosis/drug effects , Caco-2 Cells , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Interactions , ErbB Receptors/metabolism , Humans , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology , Oxidative Stress/drug effects , Oxycodone/toxicity , Signal Transduction
7.
Genes Brain Behav ; 19(5): e12609, 2020 06.
Article in English | MEDLINE | ID: mdl-31489753

ABSTRACT

Genetic factors are theorized to contribute to the substantial inter-individual variability in opioid abuse/addiction. To advance the behavioral genetics of prescription opioid abuse, our prior work identified the 129S1/SvlmJ (S1) and related 129P3/J (P3) mouse substrains, respectively, as low and high opioid-taking. Herein, we related our prior results to measures of sucrose reward/reinforcement, basal anxiety, opioid-induced place-conditioning, locomotor activity and Straub tail reaction, as well as behavioral and physiological signs of withdrawal. Substrains were also re-examined for higher-dose oxycodone and fentanyl intake under limited-access drinking procedures. S1 mice failed to acquire sucrose self-administration under various operant-conditioning procedures and exhibited lower sucrose intake in the home-cage. However, sucrose intake under limited-access procedures escalated in both substrains with repeated sucrose experience. S1 mice exhibited less spontaneous locomotor activity, as well as less opioid-induced locomotor activity and Straub tail reaction, than P3 mice and failed to exhibit an oxycodone-induced place-preference. The lack of conditioned behavior by S1 mice was unrelated to behavioral signs of withdrawal-induced negative affect or dependence severity, but might reflect high levels of basal anxiety-like behavior. Intriguingly, S1 and P3 mice initially exhibited equivalent oxycodone and fentanyl consumption in the home-cage; however opioid intake escalated only in P3 mice with repeated opioid experience. No sex differences were observed for any of our measures. These data provide additional evidence for robust differences in opioid addiction-related behaviors between P3 and S1 substrains and suggest that anxiety, learning, and/or motivational impairments might confound interpretation of operant- and place-conditioning studies employing the S1 substrain.


Subject(s)
Anxiety/genetics , Conditioning, Operant , Genotype , Opioid-Related Disorders/genetics , Analgesics, Opioid/toxicity , Animals , Anxiety/etiology , Anxiety/physiopathology , Choice Behavior , Female , Fentanyl/toxicity , Locomotion , Male , Mice , Mice, Inbred C57BL , Motivation , Opioid-Related Disorders/complications , Opioid-Related Disorders/physiopathology , Oxycodone/toxicity , Spatial Behavior
8.
Health Econ ; 28(12): 1449-1461, 2019 12.
Article in English | MEDLINE | ID: mdl-31715653

ABSTRACT

The United States is currently in the midst of the worst drug epidemic in its history, with nearly 64,000 overdose deaths in 2016. In response, pharmaceutical companies have begun introducing abuse-deterrent painkillers, pills with properties that make the drug more difficult to misuse. The first such painkiller, a reformulated version of OxyContin, was released in 2010. Previous research has found no net effect on opioid mortality, with users substituting from OxyContin toward heroin. This paper explores health effects of the reformulation beyond mortality. In particular, I show that heroin is substantially more likely to be injected than OxyContin, increasing exposure to blood-borne diseases. Exploiting variation across states in OxyContin misuse prior to the reformulation, I find relative increases in the spread of hepatitis B and C in states most likely to be affected by the reformulation. In aggregate, the estimates suggest that absent the reformulation, we would have observed approximately 76% fewer cases of hepatitis C and 53% fewer cases of hepatitis B from 2011 to 2015. I find some suggestive evidence that the reformulation also lead to increases in HIV and hepatitis A, although these findings are less robust. These findings have important implications for future policies addressing the opioid crisis.


Subject(s)
Analgesics, Opioid/administration & dosage , Opioid-Related Disorders/epidemiology , Oxycodone/administration & dosage , Prescription Drug Misuse/statistics & numerical data , Technology, Pharmaceutical/statistics & numerical data , Analgesics, Opioid/toxicity , Drug Administration Routes , Drug Overdose/epidemiology , HIV Infections/epidemiology , Hepatitis A/epidemiology , Hepatitis B/epidemiology , Hepatitis C/epidemiology , Heroin/administration & dosage , Heroin/toxicity , Humans , Opioid-Related Disorders/complications , Opioid-Related Disorders/mortality , Oxycodone/toxicity , Prescription Drug Misuse/mortality , Substance Abuse, Intravenous/complications , Substance Abuse, Intravenous/epidemiology , United States/epidemiology
9.
Eur Rev Med Pharmacol Sci ; 23(14): 6315-6320, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31364138

ABSTRACT

Opiate withdrawal-induced psychosis is an uncommon clinical manifestation. We present a 36-year-old male patient, with no prior personal or familiar psychiatric history, in treatment with several analgesic drugs (including oxycodone) for non-inflammatory chronic rachialgia. The patient is hospitalized after exhibiting psychotic symptomatology (delusions of harm and contamination, olfactory hallucinations, and aberrant behavior). This psychotic symptomatology first manifested after abruptly interrupting his prescribed oxycodone intake. It had a fluctuating course over time (alternating between lucid states and delusional ones) and eventually subsided after the prescription of antipsychotic drugs. In this case report, we describe the follow-up of the patient and discuss the influence and relevance of oxycodone withdrawal on the psychotic symptomatology.


Subject(s)
Antipsychotic Agents/therapeutic use , Oxycodone/toxicity , Psychotic Disorders/drug therapy , Substance Withdrawal Syndrome/psychology , Adult , Humans , Male , Psychotic Disorders/etiology , Treatment Outcome
10.
Regul Toxicol Pharmacol ; 108: 104433, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31362032

ABSTRACT

PF614, a novel trypsin activated abuse protection (TAAP) prodrug of oxycodone, is being studied as chronic pain analgesic with extended release and abuse resistant properties. A series of nonclinical safety studies were conducted to support PF614 introduction to clinical trials. Ames assays (PF614 and its metabolites), comet assay (PF614 ≤ 50 mg/kg/day oral gavage in rats) and micronucleus assay (PF614 ≤ 175 mg/kg/day oral gavage in rats) were negative. hERG assay IC50 for PF614 was ≥300 µM. PF614 (0.1 and 10 µM) showed a low permeability in Caco-2 cells (≤1.17 x 10-6 cm/s) and was not a P-gp or BCRP substrate or inhibitor. The mean percent unbound PF614 among all concentrations in plasma ranged from 91.2 to 98.4, 79.4 to 100, and 52.9-79.9% in rat, dog, and human, respectively. Also, PF614 was metabolically stable in rat, dog, and human hepatocytes with no metabolites identified. Safety pharmacology study in dog indicated moderately lower heart rate at ≥ 2 mg/kg oral gavage doses. Toxicity studies of PF614 in rat and dog with daily oral doses of 25 and 18 mg/kg, respectively, for 14 Days were well tolerated with favorable safety profile supporting its further clinical evaluation.


Subject(s)
Abuse-Deterrent Formulations , Analgesics, Opioid/toxicity , Oxycodone/toxicity , Prodrugs/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Caco-2 Cells , Dogs , Electrocardiography/drug effects , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mutagenicity Tests , Neoplasm Proteins/metabolism , Rats , Transcriptional Regulator ERG/metabolism , Trypsin
11.
Drug Test Anal ; 11(9): 1460-1464, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31242346

ABSTRACT

The stability of compounds in formalin solution is an important factor for drug analysis in a toxicological investigation. In this article, the authors report a complex medico-legal case involving midazolam and oxycodone. The complexity of this case comes from the fact that the body was embalmed with formalin solution before the autopsy. This technique, called thanatopraxy, allows the preservation of corpses from decomposition, the destruction of a maximal number of micro-organisms, and the presentation of the body with a natural appearance to the family. Unfortunately, when thanatopraxy is performed before the collection of biological specimens, the toxicological results are not representative of the time of the death. In addition, the interpretation of the results is difficult, because formalin can cause oxidation of xenobiotics present in the body at the time of the death, alter the pH of the tissues and dilute the compounds. To document the chemical stability of midazolam and oxycodone in formalin solution and interpret the results, a stability study was conducted for 21 days. Blood containing midazolam and oxycodone was spiked with formalin, kept at 4°C and regularly tested for both drugs. This study showed a rapid degradation of midazolam and oxycodone (85% during the first 24 hours for oxycodone). In the peripheral blood of the victim, methanol (1.31 g/L), midazolam (74ng/mL) and oxycodone (152 ng/mL) were identified. According to the stability study, the measured concentrations in formalin fixed-tissues are to be interpreted very carefully, knowing that significant degradation has occurred.


Subject(s)
Hypnotics and Sedatives/blood , Midazolam/blood , Narcotics/blood , Oxycodone/blood , Adult , Autopsy/methods , Chromatography, High Pressure Liquid , Diagnosis , Drug Monitoring , Drug Overdose/blood , Drug Overdose/diagnosis , Forensic Toxicology , Formaldehyde/chemistry , Humans , Hypnotics and Sedatives/toxicity , Male , Midazolam/toxicity , Narcotics/toxicity , Oxycodone/toxicity , Tandem Mass Spectrometry
12.
Mol Pharm ; 16(6): 2364-2375, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31018096

ABSTRACT

This study focused on formulating conjugate vaccines targeting oxycodone and heroin for technology transfer, good manufacturing practice (GMP), and clinical evaluation. Lead vaccines used the highly immunogenic carrier protein keyhole limpet hemocyanin (KLH), which poses formulation problems because of its size. To address this barrier to translation, an oxycodone-based hapten conjugated to GMP-grade subunit KLH (OXY-sKLH) and adsorbed on alum adjuvant was studied with regard to carbodiimide coupling reaction time, buffer composition, purification methods for conjugates, conjugate size, state of aggregation, and protein/alum ratio. Vaccine formulations were screened for post-immunization antibody levels and efficacy in reducing oxycodone distribution to the brain in rats. While larger conjugates were more immunogenic, their size prevented characterization of the haptenation ratio by standard analytical methods and sterilization by filtration. To address this issue, conjugation chemistry and vaccine formulation were optimized for maximal efficacy, and conjugate size was measured by dynamic light scattering prior to adsorption to alum. An analogous heroin vaccine (M-sKLH) was also optimized for conjugation chemistry, formulated in alum, and characterized for potency against heroin in rats. Finally, this study found that the efficacy of OXY-sKLH was preserved when co-administered with M-sKLH, supporting the proof of concept for a bivalent vaccine formulation targeting both heroin and oxycodone. This study suggests methods for addressing the unique formulation and characterization challenges posed by conjugating small molecules to sKLH while preserving vaccine efficacy.


Subject(s)
Opioid-Related Disorders/prevention & control , Vaccines, Conjugate/chemistry , Animals , Hemocyanins/metabolism , Heroin/toxicity , Humans , Oxycodone/toxicity , Rats , Vaccines/chemistry , Vaccines/therapeutic use , Vaccines, Conjugate/therapeutic use
14.
Drugs R D ; 16(2): 155-63, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26913723

ABSTRACT

BACKGROUND AND OBJECTIVES: Oxycodone is the mo st commonly used opioid for the treatment of moderate to severe pain. The peak cerebrospinal fluid concentration after epidural oxycodone was reported to be 300-fold greater (0.025 mM) than when administered intravenously after gynecologic surgery. Additionally, those patients administered epidural oxycodone had lower pain scores, needed less rescue analgesics and had fewer adverse effects compared with intravenous administration. However, oxycodone neurotoxicity requires evaluation before intrathecal implementation for routine clinical use. METHODS: We used two in vitro cell culture models to compare the cytotoxicity of oxycodone with that of morphine, and to study the mechanisms underlying toxicity. Human neuroblastoma cells and mouse motoneuronal cells were treated with increasing concentrations (0.0125-2 mM) of oxycodone or morphine, and were harvested at 24, 48 or 96 h. Cell cultures were evaluated with 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide and resazurin reduction assays. RESULTS: Both morphine and oxycodone decreased cell viability in a dose-dependent manner at concentrations between 0.5 and 2 mM. Morphine increased the number of apoptotic cells compared with oxycodone when assessed by flow cytometry, and transmission electron microscopy images revealed that exposure to both opioids evoked the appearance of numerous electron-dense, probable autophagic vacuoles in the cytoplasm of the cells. CONCLUSIONS: Based on these results, it seems that the cytotoxicity of oxycodone in motoneuronal cells is similar to or less than that of morphine, and occurs only at concentrations above the peak clinical concentration in the cerebrospinal fluid after epidural administration.


Subject(s)
Analgesics, Opioid/toxicity , Morphine/toxicity , Oxycodone/toxicity , Analgesics, Opioid/adverse effects , Analgesics, Opioid/chemistry , Analgesics, Opioid/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Drug Therapy, Combination , Flow Cytometry , Humans , Mice , Morphine/adverse effects , Morphine/chemistry , Morphine/therapeutic use , Motor Neurons/pathology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Neurotoxicity Syndromes/etiology , Oxycodone/adverse effects , Oxycodone/chemistry , Oxycodone/therapeutic use
15.
Environ Mol Mutagen ; 56(9): 777-87, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25913631

ABSTRACT

14-Hydroxycodeinone (14-HC) is an α,ß-unsaturated ketone impurity found in oxycodone drug substance and has a structural alert for genotoxicity. 14-HC was tested in a combined Modified and Standard Comet Assay to determine if the slight decrease in % Tail DNA noted in a previously conducted Standard Comet Assay with 14-HC could be magnified to clarify if the response was due to cross-linking activity. One limitation of the Standard Comet Assay is that DNA cross-links cannot be reliably detected. However, under certain modified testing conditions, DNA cross-links and chemical moieties that elicit such cross-links can be elucidated. One such modification involves the induction of additional breakages of DNA strands by gamma or X-ray irradiation. To determine if 14-HC is a DNA crosslinker in vivo, a Modified Comet Assay was conducted using X-ray irradiation as the modification to visualize crosslinking activity. In this assay, 14-HC was administered orally to mice up to 320 mg/kg/day. Results showed a statistically significant reduction in percent tail DNA in duodenal cells at 320 mg/kg/day, with a nonstatistically significant but dose-related reduction in percent tail DNA also observed at the mid dose of 160 mg/kg/day. Similar decreases were not observed in cells from the liver or stomach, and no increases in percent tail DNA were noted for any tissue in the concomitantly conducted Standard Comet Assay. Taken together, 14-HC was identified as a cross-linking agent in the duodenum in the Modified Comet Assay.


Subject(s)
Comet Assay/methods , Oxycodone/analogs & derivatives , Administration, Oral , Animals , Chlorambucil/toxicity , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/toxicity , DNA Damage/drug effects , DNA Damage/radiation effects , Dose-Response Relationship, Drug , Hempa/toxicity , Liver/drug effects , Liver/radiation effects , Male , Mice, Inbred ICR , Mitomycin/chemistry , Mitomycin/toxicity , Oxycodone/administration & dosage , Oxycodone/chemistry , Oxycodone/toxicity , Stomach/drug effects , X-Rays
17.
Semin Pediatr Neurol ; 21(2): 160-5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25149954

ABSTRACT

We present the case of a 14-year-old girl with a biphasic course after oxycodone ingestion. Clinically, she had a rapid return to baseline after initial ingestion and presented a week later with new-onset ballism, akathisia, and encephalopathy. Neuroimaging demonstrated bilateral globi pallidi and cerebellar lesions with a relative decrease of metabolite peaks on magnetic resonance spectroscopy. Her movement disorder was treated successfully with valproic acid and clonidine. Her cognitive functioning returned to baseline 3 months after ingestion.


Subject(s)
Akathisia, Drug-Induced , Brain Diseases/chemically induced , Dyskinesia, Drug-Induced , Oxycodone/toxicity , Adolescent , Akathisia, Drug-Induced/diagnostic imaging , Akathisia, Drug-Induced/drug therapy , Akathisia, Drug-Induced/pathology , Akathisia, Drug-Induced/physiopathology , Brain/diagnostic imaging , Brain/pathology , Brain Diseases/diagnostic imaging , Brain Diseases/pathology , Brain Diseases/physiopathology , Dyskinesia, Drug-Induced/diagnostic imaging , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/pathology , Dyskinesia, Drug-Induced/physiopathology , Female , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Movement Disorders , Tomography, X-Ray Computed
19.
J Pharmacol Exp Ther ; 339(3): 738-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21875950

ABSTRACT

Oxycodone, a semisynthetic opioid analgesic, is frequently prescribed for the management of pain. Side effects of nausea and emesis affect patient compliance and limit its therapeutic use. The present study established that an antinociceptive dose of oxycodone (15 mg/kg; oral) induces the pica response. We found sex differences in the temporal course of pica, with females having a longer duration. Opioid receptors mediated the pica response, as 1.0 mg/kg naloxone transiently attenuated and 2.0 mg/kg naloxone blocked pica. A κ-selective antagonist failed to block the response, suggesting mediation by µ opioid receptor. For further validation, we used the well established kaolin intake model to assess pica with the chemotherapeutic drug cisplatin as a positive control. Oxycodone and cisplatin significantly increased kaolin intake 4- to 7-fold, and the wet weight of stomach was elevated 2- to 3-fold. To examine the underlying neural circuitry, we investigated c-fos activation in the area postrema and nucleus of solitary tract (NTS). Oxycodone treatment significantly increased the number of c-fos-positive neurons in the area postrema and NTS compared with water controls. As expected, cisplatin also increased the number of c-fos-positive cells in these regions. In the area postrema, the oxycodone effect was greater than cisplatin, especially at 2 h. These results indicate that an antinociceptive dose of oxycodone is associated with the expression of pica, a pro-emetic response.


Subject(s)
Analgesics, Opioid/toxicity , Brain/drug effects , Nausea/drug therapy , Oxycodone/toxicity , Pica/chemically induced , Vomiting/drug therapy , Analgesics, Opioid/pharmacology , Animals , Antiemetics/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cisplatin/pharmacology , Cisplatin/toxicity , Drug Evaluation, Preclinical , Emetics/pharmacology , Female , Humans , Kaolin/metabolism , Kaolin/pharmacology , Male , Narcotic Antagonists/pharmacology , Nausea/chemically induced , Oxycodone/pharmacology , Pica/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/antagonists & inhibitors , Sex Characteristics , Time Factors , Vomiting/chemically induced
20.
Ther Drug Monit ; 32(3): 318-23, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20418801

ABSTRACT

Identification of maternal opioid abuse in pregnancy is often difficult to ascertain in the absence of a reliable self-report. We aimed to characterize an at-risk neonatal population for opioid exposures as well as other drugs of abuse and alcohol. From June 2007 to January 2009, 563 neonatal hair and 1318 meconium specimens were assessed for opioids and were positive in 11.4% and 17.0%, respectively. Neonates testing positive for opioids in hair or meconium analysis were also more likely to test positive for other licit and illicit substances (odds ratiohair, 1.75; 95% confidence interval, 1.03-2.97; odds ratiomeconium, 1.61; 95% confidence interval, 1.16-2.22). Specifically, a positive neonatal hair test for opioids also predicted a positive result for oxycodone. In addition, a positive meconium test result for opioids was associated with positive results for cocaine, oxycodone, methadone, benzodiazepines, and fatty acid ethyl esters (alcohol). Finally, there was a significant correlation between maternal and neonatal hair test results for opioids (Spearman rank rho = 0.657, P = 0.03). Understanding the addiction profiles of these women may lead to better clinical and social management and may largely benefit an at-risk population.


Subject(s)
Analgesics, Opioid/analysis , Fetus/drug effects , Hair/chemistry , Meconium/metabolism , Narcotics/toxicity , Analgesics, Opioid/toxicity , Cocaine/toxicity , Ethanol/toxicity , Fatty Acids/analysis , Female , Fetal Alcohol Spectrum Disorders/epidemiology , Fetal Alcohol Spectrum Disorders/etiology , Hair/drug effects , Humans , Infant, Newborn , Maternal-Fetal Exchange , Oxycodone/toxicity , Population Groups , Pregnancy , Pregnancy Complications/epidemiology , Risk , Substance Abuse Detection/methods , Substance-Related Disorders
SELECTION OF CITATIONS
SEARCH DETAIL
...