Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
Add more filters

Publication year range
1.
BMC Vet Res ; 18(1): 196, 2022 May 25.
Article in English | MEDLINE | ID: mdl-35614473

ABSTRACT

BACKGROUND: In humans, codeine is a commonly prescribed analgesic that produces its therapeutic effect largely through metabolism to morphine. In some species, analgesic effects of morphine have also been attributed to the morphine-6-glucuronide (M6G) metabolite. Although an effective analgesic, administration of morphine to horses produces dose-dependent neuroexcitation at therapeutic doses. Oral administration of codeine at a dose of 0.6 mg/kg has been shown to generate morphine and M6G concentrations comparable to that observed following administration of clinically effective doses of morphine, without the concomitant adverse effects observed with morphine administration. Based on these results, it was hypothesized that codeine administration would provide effective analgesia with decreased adverse excitatory effects compared to morphine. Seven horses received a single oral dose of saline or 0.3, 0.6 or 1.2 mg/kg codeine or 0.2 mg/kg morphine IV (positive control) in a randomized balanced 5-way cross-over design. Blood samples were collected up to 72 hours post administration, codeine, codeine 6-glucuronide, norcodeine morphine, morphine 3-glucuronide and M6G concentrations determined by liquid chromatography- mass spectrometry and pharmacokinetic analysis performed. Pre- and post-drug related behavior, locomotor activity, heart rate and gastrointestinal borborygmi were recorded. Response to noxious stimuli was evaluated by determining thermal threshold latency. RESULTS: Morphine concentrations were highest in the morphine dose group at all times post administration, however, M6G concentrations were significantly higher in all the codeine dose groups compared to the morphine group starting at 1 hour post drug administration and up to 72-hours in the 1.2 mg/kg group. With the exception of one horse that exhibited signs of colic following administration of 0.3 and 0.6 mg/kg, codeine administration was well tolerated. Morphine administration, led to signs of agitation, tremors and excitation. There was not a significant effect on thermal nociception in any of the dose groups studied. CONCLUSIONS: The current study describes the metabolic profile and pharmacokinetics of codeine in horses and provides information that can be utilized in the design of future studies to understand the anti-nociceptive and analgesic effects of opioids in this species with the goal of promoting judicious and safe use of this important class of drugs.


Subject(s)
Codeine , Glucuronides , Nociception , Analgesics, Opioid , Animals , Codeine/adverse effects , Codeine/pharmacokinetics , Glucuronides/adverse effects , Glucuronides/pharmacokinetics , Horses , Morphine , Morphine Derivatives/adverse effects , Morphine Derivatives/pharmacokinetics
2.
BMC Vet Res ; 17(1): 77, 2021 Feb 13.
Article in English | MEDLINE | ID: mdl-33581736

ABSTRACT

BACKGROUND: In humans, the drug metabolizing enzyme CYP2D6 is highly polymorphic resulting in substantial differences in the metabolism of drugs including anti-arrhythmics, neuroleptics, and opioids. The objective of this study was to phenotype a population of 100 horses from five different breeds and assess differences in the metabolic activity of the equine CYP2D6 homolog using codeine as a probe drug. Administration of a probe drug is a common method used for patient phenotyping in human medicine, whereby the ratio of parent drug to metabolite (metabolic ratio, MR) can be used to compare relative enzyme function between individuals. A single oral dose of codeine (0.6 mg/kg) was administered and plasma concentrations of codeine and its metabolites were determined using liquid chromatography mass spectrometry. The MR of codeine O-demethylation [(codeine)/(morphine + morphine-3-glucuronide + morphine-6-glucuronide)] was determined using the area under the plasma concentration-time curve extrapolated from time zero to infinity (AUC0-∞) for each analyte and used to group horses into predicted phenotypes (high-, moderate-, and low-MR). RESULTS: The MR of codeine O-demethylation ranged from 0.002 to 0.147 (median 0.018) among all horses. No significant difference in MR was observed between breeds, age, or sex. Of the 100 horses, 11 were classified as high-MR, 72 moderate-MR, and 17 low-MR. Codeine AUC0-∞ and O-demethylation MR were significantly different (p < 0.05) between all three groups. The mean ± SD MR was 0.089 ± 0.027, 0.022 ± 0.011, and 0.0095 ± 0.001 for high-, moderate-, and low-MR groups, respectively. The AUC for the morphine metabolites morphine-3-glucuronide and morphine-6-glucuronide were significantly different between high-and low-MR groups (p < 0.004 and p < 0.006). CONCLUSIONS: The MR calculated from plasma following codeine administration allowed for classification of horses into metabolic phenotypes within a large population. The range of codeine metabolism observed among horses suggests the presence of genetic polymorphisms in CYP2D82 of which codeine is a known substrate. Additional studies including CYP2D82 genotyping of high- and low-MR individuals are necessary to determine the presence of CYP2D polymorphisms and their functional implications with respect to the metabolism of therapeutics.


Subject(s)
Codeine/metabolism , Codeine/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Animals , Codeine/analogs & derivatives , Codeine/blood , Cytochrome P-450 CYP2D6/genetics , Female , Horses , Male , Morphine Derivatives/metabolism , Phenotype , Polymorphism, Genetic
3.
Vet Anaesth Analg ; 47(5): 694-704, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32654915

ABSTRACT

OBJECTIVE: To describe the pharmacokinetics and selected pharmacodynamic variables of codeine and its metabolites in Thoroughbred horses following a single oral administration. STUDY DESIGN: Prospective experimental study. ANIMALS: A total of 12 Thoroughbred horses, nine geldings and three mares, aged 4-8 years. METHODS: Horses were administered codeine (0.6 mg kg-1) orally and blood was collected before administration and at various times until 120 hours post administration. Plasma and urine samples were collected and analyzed for codeine and its metabolites by liquid chromatography-mass spectrometry, and plasma pharmacokinetics were determined. Heart rate and rhythm, step counts, packed cell volume and total plasma protein were measured before and 4 hours after administration. RESULTS: Codeine was rapidly converted to the metabolites norcodeine, codeine-6-glucuronide (C6G), morphine, morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G). Plasma codeine concentrations were best represented using a two-compartment model. The Cmax, tmax and elimination t½ were 270.7 ± 136.0 ng mL-1, 0.438 ± 0.156 hours and 2.00 ± 0.534 hours, respectively. M3G was the main metabolite detected (Cmax 492.7 ± 35.5 ng mL-1), followed by C6G (Cmax 96.1 ± 33.8 ng mL-1) and M6G (Cmax 22.3 ± 4.96 ng mL-1). Morphine and norcodeine were the least abundant metabolites with Cmax of 3.17 ± 0.95 and 1.42 ± 0.79 ng mL-1, respectively. No significant adverse or excitatory effects were observed. CONCLUSIONS AND CLINICAL RELEVANCE: Following oral administration, codeine is rapidly metabolized to morphine, M3G, M6G, C6G and norcodeine in horses. Plasma concentrations of M6G, a presumed active metabolite of morphine, were comparable to concentrations reported previously following administration of an analgesic dose of morphine to horses. Codeine was well tolerated based on pharmacodynamic variables and behavioral observations.


Subject(s)
Codeine/pharmacokinetics , Horses/metabolism , Administration, Oral , Animals , Area Under Curve , Codeine/blood , Codeine/metabolism , Codeine/urine , Drug Administration Schedule , Female , Half-Life , Male
4.
Biopharm Drug Dispos ; 40(9): 350-357, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31691978

ABSTRACT

We previously analysed the serum concentrations of dihydrocodeine in a 1-month-old infant with respiratory depression after being prescribed dihydrocodeine phosphate 2.0 mg/day divided t.i.d. for 2 days. The purpose was to develop a full physiologically based pharmacokinetic (PBPK) model that could account for these and other drug monitoring results. Based on experiments in Caco-2 cell monolayers, the effective permeability of dihydrocodeine in human jejunum was established as 1.28 × 10-4 cm/s. The in vitro Vmax /Km values for dihydrocodeine demethylation mediated by recombinant cytochrome P450 2D6 and 3A4 were 0.19 and 0.066 µl/min/pmol, respectively, and for dihydrocodeine 6-O-glucuronidation mediated by recombinant UGT2B4 and 2B7, the Vmax /Km values were 0.14 and 0.22 µl/min/mg protein, respectively. Renal clearance was calculated as 5.37 L/h on the total clearance value multiplied by the fraction recovered in urine. The reported plasma concentration-time profiles of dihydrocodeine after intravenous administration in healthy volunteers were used to adjust the tissue partitioning ratios. The developed model simulated the pharmacokinetic profiles of dihydrocodeine after single and multiple oral administrations reasonably well in the same population. Subsequently, the validated model was used to simulate pharmacokinetic profiles for five pediatric cases, including the 1-month-old Japanese boy and a 14-year-old Japanese girl who took an overdose of dihydrocodeine phosphate (37 mg). The simulated pharmacokinetic profiles for five virtual pediatric subjects matching the age, gender, and P450 2D6 phenotype of each case approximately reflected the observed values. These results suggested that our dihydrocodeine PBPK model reproduced the results of clinical cases reasonably well for subjects.


Subject(s)
Codeine/analogs & derivatives , Models, Biological , Administration, Intravenous , Administration, Oral , Adolescent , Adult , Analgesics, Opioid/pharmacokinetics , Codeine/administration & dosage , Codeine/pharmacokinetics , Female , Humans , Infant, Newborn , Male
5.
J Am Pharm Assoc (2003) ; 59(4): 469-473, 2019.
Article in English | MEDLINE | ID: mdl-31126828

ABSTRACT

OBJECTIVES: The objectives of this commentary are to: (1) briefly describe the ongoing challenges of addressing opioid abuse, (2) examine codeine metabolism in terms of pharmacogenomics, (3) describe 2 points of patient contact where pharmacogenomics can be implemented to determine the appropriateness of opioid therapy, and (4) briefly explore the value of pharmacogenomics in opioid-abuse and dependency research. SUMMARY: Opioid abuse is one of the most significant medical, social, and economic threats facing our country today. Yet for some patients, opioids are the only effective treatment option in achieving pain relief. Differentiating patients who are susceptible to abuse and addiction from those who are not has been absent in standard statistics-based medication prescribing. Pharmacogenomics (PGX) is a burgeoning science that examines how gene variations (variants) influence drug metabolism. With an estimated 23% of the U.S. population unable to properly metabolize codeine and related analogs, PGX could play an immediate role in the management of opioid therapy if applied (1) as part of regular pre-operative screening assessments and (2) prior to or in conjunction with pain-management referral. CONCLUSION: Using PGX to identify patients who should not begin or continue treatment with opioids because of abnormal metabolic pathways could (1) reduce the number of opioid prescriptions written; (2) reduce related costs; (3) guide more patient-centric, pain-management treatment plans, away from opioids as necessary; and (4) reduce the potential for opioid abuse and addiction.


Subject(s)
Analgesics, Opioid/administration & dosage , Opioid-Related Disorders/prevention & control , Pain/drug therapy , Pharmacogenetics , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Codeine/administration & dosage , Codeine/adverse effects , Codeine/pharmacokinetics , Humans , Opioid-Related Disorders/genetics
6.
AAPS PharmSciTech ; 19(7): 3057-3066, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30091062

ABSTRACT

The objective of this study was to prepare ibuprofen enteric-coated sustained-release pellets (IB-SRPs) and codeine phosphate immediate-release pellets (CP-IRPs) to play a synergistic role in analgesia. The pellets were developed by extrusion-spheronization and fluidized bed coating technology. The single-factor investigation was used to determine the optimal prescription and process. The sustained-release membrane of IB-SRPs was water-insoluble ethyl cellulose (EC), triethyl citrate (TEC) was used as plasticizer, and hydroxypropyl methylcellulose (HPMCP) was chose as porogen. Besides, the immediate-release layer of CP-IRPs was gastric-soluble coating film. The ibuprofen and codeine phosphate compound capsules (IB-CP SRCs) were prepared by IB-SRPs and CP-IRPs packed together in capsules with the optimum doses of 200 and 13 mg, respectively. The prepared pellets were evaluated by scanning electron microscopy and dissolution test. Pharmacokinetic studies in beagle dogs indicated that the optimized IB-CP SRCs had smaller individual differences and better reproducibility comparing with commercial available tablets. Additionally, IB-CP SRCs achieved consistency with in vivo and in vitro tests. Therefore, IB-CP SRCs could play a great role in rapid and long-term analgesic.


Subject(s)
Codeine/chemical synthesis , Codeine/pharmacokinetics , Ibuprofen/chemical synthesis , Ibuprofen/pharmacokinetics , Animals , Capsules , Delayed-Action Preparations/chemical synthesis , Delayed-Action Preparations/pharmacokinetics , Dogs , Drug Evaluation, Preclinical/methods , Drug Implants/chemical synthesis , Drug Implants/pharmacokinetics , Reproducibility of Results , Solubility , Tablets, Enteric-Coated
7.
Ther Drug Monit ; 39(1): 88-90, 2017 02.
Article in English | MEDLINE | ID: mdl-28081042

ABSTRACT

A retrospective analysis of oral fluid drug testing results using LC-MS/MS was performed to determine the prevalence rates in oral fluid for codeine (COD) and 3 COD metabolites-morphine (MOR), norhydrocodone (NHC), and hydrocodone (HCOD). Oral fluid samples were collected using Quantisal oral fluid collection device (Immunalysis Inc.) and submitted to Millennium Health, LLC for the routine drug analysis by LC-MS/MS. Consistent with previously published literature, COD was the primary analyte detected in oral fluid after the use of COD. In COD-positive samples, HCOD, MOR, and NHC were detected at rates of 68.4%, 18.4%, and 6.3%, respectively. Concentration ranges of these analytes were 1.0 to >2000 ng/mL for COD, 1.0-20.2 ng/mL for MOR, 1.0-740.0 ng/mL for HCOD, and 2.1-47.5 ng/mL for NHC. In contrast to urine, where HCOD is typically detected as a minor metabolite of COD, HCOD was the most commonly detected metabolite in oral fluid in samples testing positive for COD with reported prescriptions for COD. This observation suggests that care should be taken when interpreting HCOD positives in oral fluid results, and that the use of COD should be considered as one possible explanation for HCOD positives.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Chromatography, Liquid/methods , Codeine/pharmacokinetics , Tandem Mass Spectrometry/methods , Analgesics, Opioid/administration & dosage , Codeine/administration & dosage , Humans , Hydrocodone/analogs & derivatives , Hydrocodone/analysis , Hydrocodone/metabolism , Morphine/analysis , Morphine/metabolism , Retrospective Studies , Substance Abuse Detection/methods
8.
Biopharm Drug Dispos ; 38(3): 231-250, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27977852

ABSTRACT

Merits of the segregated flow model (SFM), highlighting the intestine as inert serosa and active enterocyte regions, with a smaller fractional (fQ < 0.3) intestinal flow (QI ) perfusing the enterocyte region, are described. Less drug in the circulation reaches the enterocytes due to the lower flow (fQ QI ) in comparison with drug administered into the gut lumen, fostering the idea of route-dependent intestinal removal. The SFM has been found superior to the traditional model (TM), which views the serosa and enterocytes totally as a well-mixed tissue perfused by 100% of the intestinal flow, QI . The SFM model is able to explain the lower extents of intestinal metabolism of enalapril, morphine and midazolam with i.v. vs. p.o. dosing. For morphine, the urine/bile ratio of the metabolite, morphine glucuronide MGurineMGbile for p.o. was 2.6× that of i.v. This was due to the higher proportion of intestinally formed morphine glucuronide, appearing more in urine than in bile due to its low permeability and greater extent of intestinal formation with p.o. administration. By contrast, the TM predicted the same MGurineMGbile for p.o. vs. i.v. The TM predicted that the contributions of the intestine:liver to first-pass removal were 46%:54% for both p.o. and i.v. The SFM predicted same 46%:54% (intestine:liver) for p.o., but 9%:91% for i.v. By contrast, the kinetics of codeine, the precursor of morphine, was described equally well by the SFM- and TM-PBPK models, a trend suggesting that intestinal metabolism of codeine is negligible. Fits to these PBPK models further provide insightful information towards metabolite formation: available fractions and the fractions of hepatic and total clearances that form the metabolite in question. The SFM-PBPK model is useful to identify not only the presence of intestinal metabolism but the contributions of the intestine and liver for metabolite formation. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Codeine/pharmacokinetics , Intestinal Mucosa/metabolism , Liver/metabolism , Models, Biological , Morphine/pharmacokinetics , Administration, Intravenous , Administration, Oral , Animals , Bile/metabolism , Codeine/administration & dosage , Humans , Morphine/administration & dosage
9.
Biopharm Drug Dispos ; 38(1): 50-74, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27925239

ABSTRACT

The physiologically based model with segregated flow to the intestine (SFM-PBPK; partial, lower flow to enterocyte region vs. greater flow to serosal region) was found to describe the first-pass glucuronidation of morphine (M) to morphine-3ß-glucuronide (MG) in rats after intraduodenal (i.d.) and intravenous (i.v.) administration better than the traditional model (TM), for which a single intestinal flow perfused the whole of the intestinal tissue. The segregated flow model (SFM) described a disproportionately greater extent of intestinal morphine glucuronidation for i.d. vs. i.v. administration. The present study applied the same PBPK modeling approaches to examine the contributions of the intestine and liver on the first-pass metabolism of the precursor, codeine (C, 3-methylmorphine) in the rat. Unexpectedly, the profiles of codeine, morphine and morphine-3ß-glucuronide in whole blood, bile and urine, assayed by LCMS, were equally well described by both the TM-PBPK and SFM-PBPK. The fitted parameters for the models were similar, and the net formation intrinsic clearance of morphine (from codeine) for the liver was much higher, being 9- to 13-fold that of the intestine. Simulations, based on the absence of intestinal formation of morphine, correlated well with observations. The lack of discrimination of SFM and TM with the codeine data did not invalidate the SFM-PBPK model but rather suggests that the liver is the only major organ for codeine metabolism. Because of little or no contribution by the intestine to the metabolism of codeine, both the TM- and SFM-PBPK models are equally consistent with the data. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Intestinal Mucosa/metabolism , Models, Biological , Analgesics, Opioid/blood , Analgesics, Opioid/urine , Animals , Bile/metabolism , Codeine/blood , Codeine/urine , Intestinal Absorption , Liver/metabolism , Male , Morphine/blood , Morphine/urine , Morphine Derivatives/blood , Morphine Derivatives/urine , Rats, Sprague-Dawley
10.
Appl Nurs Res ; 38: 107-110, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29241501

ABSTRACT

PURPOSE: When codeine and tramadol are used for pain management, it is imperative that nurses are able to assess for potential drug-gene and drug-drug-gene interactions that could adversely impact drug metabolism and ultimately pain relief. Both drugs are metabolized through the CYP2D6 metabolic pathway which can be affected by medications as well the patient's own pharmacogenotype. The purpose of this brief report is to identify drug-gene and drug-drug-gene interactions in 30 adult patients prescribed codeine or tramadol for pain. METHODS: We used three data sources: (1) six months of electronic health record data on the number and types of medications prescribed to each patient; (2) each patient's CYP2D6 pharmacogenotype, and (3) published data on known CYP2D6 gene-drug and drug-drug-gene interactions. RESULTS: Ten patients (33%) had possible drug-gene or drug-drug-gene interactions. Five patients had CYP2D6 drug-gene interactions indicating they were not good candidates for codeine or tramadol. In addition, five patients had potential CYP2D6 drug-drug-gene interactions with either codeine or tramadol. CONCLUSION: Our findings from this exploratory study underscores the importance of assessing and accounting for drug-gene and drug-drug-gene interactions in patients prescribed codeine or tramadol.


Subject(s)
Analgesics, Opioid/therapeutic use , Codeine/therapeutic use , Cytochrome P-450 CYP2D6/genetics , Pharmacogenetics , Tramadol/therapeutic use , Adult , Aged , Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Female , Humans , Male , Middle Aged , Tramadol/pharmacokinetics , Young Adult
11.
Drug Dev Ind Pharm ; 42(6): 862-870, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26227813

ABSTRACT

CONTEXT: Conventional pain management approaches have limitations such as gastrointestinal side effects, frequent dosing, and difficulties in swallowing medications. Hence, to overcome these limitations, we developed a transdermal analgesic patch. OBJECTIVE: This study was designed to formulate a drug in adhesive transdermal patch with codeine (CDB) and acetaminophen (APAP) that may potentially treat moderate pain in children. MATERIALS AND METHODS: Three analgesic drugs hydrocodone bitartrate, CDB and APAP were screened by a slide crystallization study using polarized light microscope and their permeation profiles were studied using vertical Franz diffusion cells across porcine ear skin, dermatomed human skin and epidermis for 24 h, and the samples were quantified by high performance liquid chromatography. Patches used for permeation studies were prepared by dissolving sub-saturation concentration of the drug(s) in adhesive (with/without 5% w/w oleic acid [OA]), cast with a film casting knife. RESULTS AND DISCUSSION: Among the three drugs screened, CDB demonstrated the best permeation profile (660.21 µg/cm2), and shortest lag time (4.35 ± 0.01 h), and hence was chosen for patch studies. The highest concentration of CDB in the patch at which drug does not crystallize was determined as 40% of its saturation solubility (Cs) and that of APAP was determined as 200% of its Cs. CDB standalone patch delivered 105.48 µg/cm2 of CDB, while the CDB-APAP combination patch with 5% w/w OA delivered 151.40 µg/cm2 CDB and 58.12 µg/cm2 APAP in 24 h. CONCLUSION: Drug-in-adhesive patches using CDB and APAP were developed for infants and children. Addition of OA enhanced solubility and permeation of drugs.


Subject(s)
Acetaminophen/chemistry , Analgesics, Non-Narcotic/chemistry , Analgesics, Opioid/chemistry , Codeine/chemistry , Pain Management/methods , Transdermal Patch , Acetaminophen/administration & dosage , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/pharmacokinetics , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Cadaver , Calorimetry, Differential Scanning , Child , Child, Preschool , Codeine/administration & dosage , Codeine/pharmacokinetics , Crystallization , Drug Combinations , Drug Compounding/methods , Epidermis/metabolism , Humans , Hydrocodone/administration & dosage , Hydrocodone/chemistry , Hydrocodone/pharmacokinetics , Infant , Permeability , Solubility , Swine
12.
J Vet Pharmacol Ther ; 39(5): 514-7, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26896302

ABSTRACT

The purpose of this study was to determine the pharmacokinetic and antinociceptive effects of an acetaminophen/codeine combination administered orally to six healthy greyhounds. Antinociception was assessed using an electronic von Frey (vF) device as a mechanical/pressure model. Acetaminophen was administered at a dose of 600 mg (14.4-23.1 mg/kg) and codeine phosphate at 90 mg (2.1-3.3 mg/kg) equivalent to 67.5 mg codeine base (1.6-2.5 mg/kg). The geometric mean maximum plasma concentrations of acetaminophen, codeine, and codeine-6-glucuronide were 7.95 µg/mL, 11.0 ng/mL, and 3819 ng/mL, respectively. Morphine concentrations were <1 ng/mL. The terminal half-lives of acetaminophen, codeine, and codeine-6-glucuronide were 0.94, 1.71, and 3.12 h. There were no significant changes in vF thresholds, except at 12 h which decreased on average by 17% compared to baseline. The decrease in vF thresholds at 12 h could be due to aversion, hyperalgesia, or random variability. The lack of antinociception in this study could be due to a true lack of antinociception, lack of model sensitivity, or specificity. Further studies using different models (including clinical trials), different dog breeds, multiple dose regimens, and a range of dosages are needed prior to recommended use or concluding lack of efficacy for oral acetaminophen/codeine in dogs.


Subject(s)
Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Acetaminophen/administration & dosage , Acetaminophen/blood , Acetaminophen/pharmacology , Administration, Oral , Analgesics, Non-Narcotic/administration & dosage , Analgesics, Non-Narcotic/blood , Analgesics, Non-Narcotic/pharmacology , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/blood , Analgesics, Opioid/pharmacology , Animals , Codeine/administration & dosage , Codeine/analogs & derivatives , Codeine/blood , Codeine/pharmacology , Dogs/metabolism , Drug Combinations , Pain/drug therapy
13.
Schmerz ; 30(6): 519-525, 2016 Dec.
Article in German | MEDLINE | ID: mdl-27439327

ABSTRACT

Opioids are an important component of the drug treatment of patients with acute and chronic pain. They differ in effectiveness, side effect profile and the risk of interactions. In this article the pharmacokinetic mechanisms of drug-drug interactions at the level of biotransformation are described and the clinical consequences which can arise are discussed. The relation of the active components to the two isoenzymes CYP2D6 and CYP3A4 is of major importance for assessing the potential drug-drug interactions of opioid analgesics at the level of the cytochrome P450 enzyme.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Analgesics, Opioid/therapeutic use , Pain/drug therapy , Aged , Analgesics, Opioid/adverse effects , Biotransformation , Codeine/adverse effects , Codeine/pharmacokinetics , Codeine/therapeutic use , Cytochrome P-450 CYP2D6/physiology , Cytochrome P-450 CYP3A/physiology , Cytochrome P-450 Enzyme System/physiology , Dose-Response Relationship, Drug , Drug Interactions , Female , Fentanyl/adverse effects , Fentanyl/pharmacokinetics , Fentanyl/therapeutic use , Humans , Infant, Newborn , Male , Pain/blood
14.
Eur J Clin Pharmacol ; 70(1): 57-63, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24077935

ABSTRACT

PURPOSE: Codeine is an analgesic drug acting on µ-opioid receptors predominantly via its metabolite morphine formed almost exclusively by CYP2D6. Genetic polymorphisms in CYP2D6 are associated with diminished pain relief and/or severe opioid side effects. In Chinese individuals, CYP2D6*10 is the most common allele with reduced enzyme activity. In this study, we investigated the effect of this allele on the pharmacokinetics of codeine and its metabolites. METHOD: A blood sample was collected from healthy Mongolian volunteers for CYP2D6 genotyping using a PCR-RFLP assay. A pharmacokinetic study was then carried out in three groups with CYP2D6*1/*1 (n=10), CYP2D6*1/*10 (n=10) and CYP2D6*10/*10 (n=9) genotypes by collecting serial blood samples for determination of plasma levels of codeine and its metabolites, morphine, morphine 3-glucuronide (M3G) and morphine 6-glucuronide (M6G) before and after a single 30-mg oral dose of codeine phosphate. Codeine and its metabolites were measured by LC-MS/MS. RESULTS: No significant differences were observed in the pharmacokinetic parameters of codeine in the three genotype groups. However, the C( max) and AUC(0-∞) of morphine, M3G and M6G were significantly different between the study groups (P<0.05). Compared with the *1/*1 group, the AUC(0-∞) for morphine in the *1/*10 and *10/*10 groups decreased by ratios (95 % CI) of 0.93 (0.26-1.59) and 0.494 (0.135-0.853) respectively. Corresponding ratios for M3G were 0.791 (0.294-1.288) and 0.615 (0.412-0.818) and for M6G were 0.643 (0.39-0.957) and 0.423 (0.267-0.579). CONCLUSION: This study demonstrates that the CYP2D6*10 allele plays an important role in the pharmacokinetics of the O-demethylated metabolites of codeine after oral administration.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Administration, Oral , Adult , Alleles , Analgesics, Opioid/blood , Area Under Curve , Asian People/genetics , Codeine/blood , Cytochrome P-450 CYP2D6/metabolism , Female , Genotype , Humans , Male , Mongolia , Morphine/blood , Morphine Derivatives/blood , Polymorphism, Genetic , Young Adult
15.
Therapie ; 68(2): 85-91, 2013.
Article in French | MEDLINE | ID: mdl-23773349

ABSTRACT

Pholcodine is an opioid that has been widely used worldwide since 1950 for the treatment of non-productive cough in children and adults. The results of early preclinical studies but also those of recent clinical trials have shown the antitussive efficacy of pholcodine to be superior to that of codeine, of longer duration, and with an equivalent or safer toxicity profile. Also, there is no risk of addiction. Concern had been raised over a possible cross-sensitisation with neuromuscular blocking agents. While a recent assessment of the available data by the European Medicines Agency (EMA) has confirmed the favourable risk-benefit ratio of pholcodine, further studies are needed to clear this point.


Subject(s)
Antitussive Agents/therapeutic use , Codeine/analogs & derivatives , Cough/drug therapy , Morpholines/therapeutic use , Adult , Antitussive Agents/adverse effects , Child , Codeine/adverse effects , Codeine/pharmacokinetics , Codeine/therapeutic use , Codeine/toxicity , Drug Hypersensitivity , Drug Interactions , Humans , Morpholines/adverse effects , Morpholines/pharmacokinetics , Morpholines/toxicity , Neuromuscular Blocking Agents/adverse effects
16.
Am J Physiol Heart Circ Physiol ; 302(3): H582-93, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22081706

ABSTRACT

Our laboratory has shown that λ-carrageenan-induced peripheral inflammatory pain (CIP) can alter tight junction (TJ) protein expression and/or assembly leading to changes in blood-brain barrier xenobiotic permeability. However, the role of reactive oxygen species (ROS) and subsequent oxidative stress during CIP is unknown. ROS (i.e., superoxide) are known to cause cellular damage in response to pain/inflammation. Therefore, we examined oxidative stress-associated effects at the blood-brain barrier (BBB) in CIP rats. During CIP, increased staining of nitrosylated proteins was detected in hind paw tissue and enhanced presence of protein adducts containing 3-nitrotyrosine occurred at two molecular weights (i.e., 85 and 44 kDa) in brain microvessels. Tempol, a pharmacological ROS scavenger, attenuated formation of 3-nitrotyrosine-containing proteins in both the hind paw and in brain microvessels when administered 10 min before footpad injection of λ-carrageenan. Similarly, CIP increased 4-hydroxynoneal staining in brain microvessels and this effect was reduced by tempol. Brain permeability to [(14)C]sucrose and [(3)H]codeine was increased, and oligomeric assemblies of occludin, a critical TJ protein, were altered after 3 h CIP. Tempol attenuated both [(14)C]sucrose and [(3)H]codeine brain uptake as well as protected occludin oligomers from disruption in CIP animals, suggesting that ROS production/oxidative stress is involved in modulating BBB functional integrity during pain/inflammation. Interestingly, tempol administration reduced codeine analgesia in CIP animals, indicating that oxidative stress during pain/inflammation may affect opioid delivery to the brain and subsequent efficacy. Taken together, our data show for the first time that ROS pharmacological scavenging is a viable approach for maintaining BBB integrity and controlling central nervous system drug delivery during acute inflammatory pain.


Subject(s)
Blood-Brain Barrier , Capillary Permeability/drug effects , Cyclic N-Oxides/pharmacology , Membrane Proteins/metabolism , Neuralgia , Xenobiotics/pharmacokinetics , Acute Disease , Aldehydes/pharmacokinetics , Analgesics, Opioid/pharmacokinetics , Animals , Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Capillary Permeability/immunology , Carbon Radioisotopes , Codeine/pharmacokinetics , Cysteine Proteinase Inhibitors/pharmacokinetics , Hyperalgesia/drug therapy , Hyperalgesia/immunology , Hyperalgesia/metabolism , Male , Membrane Proteins/immunology , Neuralgia/drug therapy , Neuralgia/immunology , Neuralgia/metabolism , Neuritis/drug therapy , Neuritis/immunology , Neuritis/metabolism , Occludin , Oxidative Stress/immunology , Rats , Rats, Sprague-Dawley , Spin Labels , Sucrose/pharmacokinetics , Tight Junctions/drug effects , Tight Junctions/immunology , Tight Junctions/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Br J Clin Pharmacol ; 73(5): 786-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22092298

ABSTRACT

AIMS: To compare the O-demethylation (CYP2D6-mediated), N-demethylation (CYP3A4-mediated) and 6-glucuronidation (UGT2B4/7-mediated) metabolism of codeine between methadone- and buprenorphine-maintained CYP2D6 extensive metabolizer subjects. METHODS: Ten methadone- and eight buprenorphine-maintained subjects received a single 60 mg dose of codeine phosphate. Blood was collected at 3 h and urine over 6 h and assayed for codeine, norcodeine, morphine, morphine-3- and -6-glucuronides and codeine-6-glucuronide. RESULTS: The urinary metabolic ratio for O-demethylation was significantly higher (P= 0.0044) in the subjects taking methadone (mean ± SD, 2.8 ± 3.1) compared with those taking buprenorphine (0.60 ± 0.43), likewise for 6-glucuronide formation (0.31 ± 0.24 vs. 0.053 ± 0.027; P < 0.0002), but there was no significant difference (P= 0.36) in N-demethylation. Similar changes in plasma metabolic ratios were also found. In plasma, compared with those maintained on buprenorphine, the methadone-maintained subjects had increased codeine and norcodeine concentrations (P < 0.004), similar morphine (P= 0.72) and lower morphine-3- and -6- and codeine-6-glucuronide concentrations (P < 0.008). CONCLUSION: Methadone is associated with inhibition of CYP2D6 and UGTs 2B4 and 2B7 reactions in vivo, even though it is not a substrate for these enzymes. Plasma morphine was not altered, owing to the opposing effects of inhibition of both formation and elimination; however, morphine-6-glucuronide (analgesically active) concentrations were substantially reduced. Drug interactions with methadone are likely to include drugs metabolized by various UGTs and CYP2D6.


Subject(s)
Analgesics, Opioid/pharmacology , Buprenorphine/pharmacology , Codeine/pharmacokinetics , Cytochrome P-450 CYP2D6 Inhibitors , Glucuronosyltransferase/antagonists & inhibitors , Methadone/pharmacology , Adolescent , Adult , Drug Interactions , Female , Genotype , Humans , Male , Middle Aged , Statistics as Topic , Young Adult
18.
Am J Hematol ; 87(5): 534-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22430884

ABSTRACT

Because no frequency data are available for the main opiate-related polymorphisms in sickle-cell disease (SCD) populations, we decided to perform such a genotyping in a cohort of 139 individuals. For pharmacodynamics,the OPRM1 A118G and the COMT G322A single nucleotide polymorphisms (SNPs) were chosen for their negative effects on the m receptors [1,2]. For pharmacokinetics [3], important SNPs for the CYP2D6 gene (codeine to morphine conversion) and for three genes involved in morphine elimination (namely CYP3A, UGT2B7, and ABCB1) were genotyped. The allelic frequencies of the OPRM1 and COMT SNPs appeared very low (0.01 to 0.05-no double mutant homozygous),as well as the proportion of CYP2D6 poor metabolizers (1.4%)and CYP3A wild-type (17.9%) which are associated with a low morphine exposure. On the contrary, up to 35% of SCD patients may have unfavorable ABCB1 and UGT2B7 genotypes for a good morphine exposure.Obviously, pharmacokinetic studies with precise phenotype/genotype correlations are required to draw definitive conclusions.


Subject(s)
Anemia, Sickle Cell/genetics , Narcotics/pharmacokinetics , Polymorphism, Single Nucleotide , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Anemia, Sickle Cell/epidemiology , Biological Availability , Biotransformation/genetics , Black People/genetics , Catechol O-Methyltransferase/genetics , Codeine/pharmacokinetics , Cohort Studies , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Drug Resistance/genetics , Genetic Testing , Genotype , Glucuronosyltransferase/genetics , Humans , Morphine/pharmacokinetics , Receptors, Opioid, mu/genetics , White People/genetics
19.
J Pharmacokinet Pharmacodyn ; 39(6): 711-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23179857

ABSTRACT

Conventional mammillary models are frequently used for pharmacokinetic (PK) analysis when only blood or plasma data are available. Such models depend on the quality of the drug disposition data and have vague biological features. An alternative minimal-physiologically-based PK (minimal-PBPK) modeling approach is proposed which inherits and lumps major physiologic attributes from whole-body PBPK models. The body and model are represented as actual blood and tissue (usually total body weight) volumes, fractions (f ( d )) of cardiac output with Fick's Law of Perfusion, tissue/blood partitioning (K ( p )), and systemic or intrinsic clearance. Analyzing only blood or plasma concentrations versus time, the minimal-PBPK models parsimoniously generate physiologically-relevant PK parameters which are more easily interpreted than those from mammillary models. The minimal-PBPK models were applied to four types of therapeutic agents and conditions. The models well captured the human PK profiles of 22 selected beta-lactam antibiotics allowing comparison of fitted and calculated K ( p ) values. Adding a classical hepatic compartment with hepatic blood flow allowed joint fitting of oral and intravenous (IV) data for four hepatic elimination drugs (dihydrocodeine, verapamil, repaglinide, midazolam) providing separate estimates of hepatic intrinsic clearance, non-hepatic clearance, and pre-hepatic bioavailability. The basic model was integrated with allometric scaling principles to simultaneously describe moxifloxacin PK in five species with common K ( p ) and f ( d ) values. A basic model assigning clearance to the tissue compartment well characterized plasma concentrations of six monoclonal antibodies in human subjects, providing good concordance of predictions with expected tissue kinetics. The proposed minimal-PBPK modeling approach offers an alternative and more rational basis for assessing PK than compartmental models.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Models, Biological , beta-Lactams/pharmacokinetics , Antibodies, Monoclonal/blood , Biological Availability , Carbamates/pharmacokinetics , Codeine/analogs & derivatives , Codeine/pharmacokinetics , Humans , Kinetics , Liver/metabolism , Midazolam/pharmacokinetics , Piperidines/pharmacokinetics , Regional Blood Flow , Verapamil/pharmacokinetics
20.
Drug Metab Dispos ; 39(8): 1324-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21551257

ABSTRACT

In this study, the selectivity of UDP-glucuronosyltransferase (UGT) enzyme inhibition by ketamine (KTM) and the kinetics of KTM inhibition of human liver microsomal morphine (MOR) and codeine (COD) glucuronidation were characterized to explore a pharmacokinetic basis for the KTM-opioid interaction. With the exception of UGT1A4, KTM inhibited the activities of recombinant human UGT enzymes in a concentration-dependent manner. However, IC(50) values were <100 µM only for UGT2B4, UGT2B7, and UGT2B15. UGT2B7 catalyzes MOR 3- and 6-glucuronidation and the 6-glucuronidation of COD, with an additional substantial contribution of UGT2B4 to the latter reaction. Consistent with the effects of KTM on the activities of recombinant UGT2B enzyme activities, KTM competitively inhibited human liver microsomal MOR and COD glucuronidation. K(i) values for KTM inhibition of MOR 3- and 6-glucuronidation and COD 6-glucuronidation by human liver microsomes supplemented with 2% bovine serum albumin were 5.8 ± 0.1, 4.6 ± 0.2, and 3.5 ± 0.1 µM, respectively. Based on the derived inhibitor constants, in vitro-in vivo extrapolation was used to predict the effects of anesthetic and analgesic doses of KTM on MOR and COD clearances. Potentially clinically significant interactions (>50% increases in the in vivo area under the curve ratios) with MOR and COD were predicted for anesthetic doses of KTM and for a subanesthetic dose of KTM on COD glucuronidation.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Codeine/pharmacokinetics , Glucuronosyltransferase/antagonists & inhibitors , Ketamine/pharmacology , Microsomes, Liver/drug effects , Morphine/pharmacokinetics , Analgesics, Opioid/metabolism , Animals , Cell Culture Techniques , Codeine/metabolism , Culture Media , Dose-Response Relationship, Drug , Drug Interactions , Glucuronosyltransferase/genetics , HEK293 Cells , Humans , Inactivation, Metabolic , Insecta , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Morphine/metabolism , Predictive Value of Tests , Protein Binding , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL