1
|
Myers AM, Wallin CM, Richardson LM, Duran J, Neole SR, Kulaglic N, Davidson C, Perrine SA, Bowen SE, Brummelte S. The effects of buprenorphine and morphine during pregnancy: Impact of exposure length on maternal brain, behavior, and offspring neurodevelopment. Neuropharmacology 2024; 257:110060. [PMID: 38960134 PMCID: PMC11285462 DOI: 10.1016/j.neuropharm.2024.110060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/21/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
The escalating incidence of opioid-related issues among pregnant women in the United States underscores the critical necessity to understand the effects of opioid use and Medication for Opioid Use Disorders (MOUDs) during pregnancy. This research employed a translational rodent model to examine the impact of gestational exposure to buprenorphine (BUP) or morphine on maternal behaviors and offspring well-being. Female rats received BUP or morphine before conception, representing established use, with exposure continuing until postnatal day 2 or discontinued on gestational day 19 to mimic treatment cessation before birth. Maternal behaviors - including care, pup retrieval, and preference - as well as hunting behaviors and brain neurotransmitter levels were assessed. Offspring were evaluated for mortality, weight, length, milk bands, surface righting latency, withdrawal symptoms, and brain neurotransmitter levels. Our results reveal that regardless of exposure length (i.e., continued or discontinued), BUP resulted in reduced maternal care in contrast to morphine-exposed and control dams. Opioid exposure altered brain monoamine levels in the dams and offspring, and was associated with increased neonatal mortality, reduced offspring weight, and elevated withdrawal symptoms compared to controls. These findings underscore BUP's potential disruption of maternal care, contributing to increased pup mortality and altered neurodevelopmental outcomes in the offspring. This study calls for more comprehensive research into prenatal BUP exposure effects on the maternal brain and infant development with the aim to mitigate adverse outcomes in humans exposed to opioids during pregnancy.
Collapse
Affiliation(s)
- Abigail M Myers
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Chela M Wallin
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | | | - Jecenia Duran
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Surbhi R Neole
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Nejra Kulaglic
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Cameron Davidson
- Dept of Behavioral Neuroscience and Psychiatry, Wayne State University, Detroit, MI, 48202, USA
| | - Shane A Perrine
- Dept of Behavioral Neuroscience and Psychiatry, Wayne State University, Detroit, MI, 48202, USA; Translational Neuroscience Program, Wayne State University, Detroit, MI, 48202, USA
| | - Scott E Bowen
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, MI, 48202, USA; Translational Neuroscience Program, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
2
|
Pande LJ, Arnet RE, Piper BJ. An Examination of the Complex Pharmacological Properties of the Non-Selective Opioid Modulator Buprenorphine. Pharmaceuticals (Basel) 2023; 16:1397. [PMID: 37895868 PMCID: PMC10610465 DOI: 10.3390/ph16101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The goal of this review is to provide a recent examination of the pharmacodynamics as well as pharmacokinetics, misuse potential, toxicology, and prenatal consequences of buprenorphine. Buprenorphine is currently a Schedule III opioid in the US used for opioid-use disorder (OUD) and as an analgesic. Buprenorphine has high affinity for the mu-opioid receptor (MOR), delta (DOR), and kappa (KOR) and intermediate affinity for the nociceptin (NOR). Buprenorphine's active metabolite, norbuprenorphine, crosses the blood-brain barrier, is a potent metabolite that attenuates the analgesic effects of buprenorphine due to binding to NOR, and is responsible for the respiratory depressant effects. The area under the concentration curves are very similar for buprenorphine and norbuprenorphine, which indicates that it is important to consider this metabolite. Crowding sourcing has identified a buprenorphine street value (USD 3.95/mg), indicating some non-medical use. There have also been eleven-thousand reports involving buprenorphine and minors (age < 19) at US poison control centers. Prenatal exposure to clinically relevant dosages in rats produces reductions in myelin and increases in depression-like behavior. In conclusion, the pharmacology of this OUD pharmacotherapy including the consequences of prenatal buprenorphine exposure in humans and experimental animals should continue to be carefully evaluated.
Collapse
Affiliation(s)
- Leana J. Pande
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Touro College of Osteopathic Medicine, Middletown, NY 10027, USA
| | - Rhudjerry E. Arnet
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
| | - Brian J. Piper
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Center for Pharmacy Innovation and Outcomes, Danville, PA 17821, USA
| |
Collapse
|
3
|
Tobacyk J, Parks BJ, Salazar P, Coward LU, Berquist MD, Gorman GS, Brents LK. Interaction between buprenorphine and norbuprenorphine in neonatal opioid withdrawal syndrome. Drug Alcohol Depend 2023; 249:110832. [PMID: 37385117 PMCID: PMC10573081 DOI: 10.1016/j.drugalcdep.2023.110832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023]
Abstract
Buprenorphine (BUP) is the preferred treatment for opioid use disorder during pregnancy but can cause neonatal opioid withdrawal syndrome (NOWS). Norbuprenorphine (NorBUP), an active metabolite of BUP, is implicated in BUP-associated NOWS. We hypothesized that BUP, a low-efficacy agonist of mu opioid receptors, will not antagonize NorBUP, a high-efficacy agonist of mu opioid receptors, in producing NOWS. To test this hypothesis, we treated pregnant Long-Evans rats with BUP (0, 0.01, 0.1 or 1mg/kg/day) ± NorBUP (1mg/kg/day) from gestation day 9 until pup delivery, and tested pups for opioid dependence using our established NOWS model. We used LC-MS-MS to quantify brain concentrations of BUP, NorBUP, and their glucuronide conjugates. BUP had little effect on NorBUP-induced NOWS, with the exception of 1mg/kg/day BUP significantly increasing NorBUP-induced NOWS by 58% in females. BUP and NorBUP brain concentrations predicted NOWS in multiple linear regression models. Interestingly, NorBUP contributed more to NOWS in females (βNorBUP = 51.34, p = 0.0001) than in males (βNorBUP = 19.21, P = 0.093), while BUP was similar for females (βBUP = 10.62, P = 0.0017) and males (βBUP = 11.38, P = 0.009). We are the first to report that NorBUP induces NOWS in the presence of BUP and it is more influential in females than males in the contribution of NorBUP to BUP-associated NOWS. These findings suggest that females are more susceptible to NorBUP-induced NOWS, and that treatment strategies that reduce prenatal NorBUP exposure may be more effective for females than males.
Collapse
Affiliation(s)
- Julia Tobacyk
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR72205, USA
| | - Brian J Parks
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR72205, USA
| | - Paloma Salazar
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR72205, USA
| | - Lori U Coward
- Department of Pharmaceutical Sciences in the McWhorter School of Pharmacy at Samford University, 800 Lakeshore Dr, Birmingham, AL35229, USA
| | - Michael D Berquist
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR72205, USA
| | - Gregory S Gorman
- Department of Pharmaceutical Sciences in the McWhorter School of Pharmacy at Samford University, 800 Lakeshore Dr, Birmingham, AL35229, USA
| | - Lisa K Brents
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR72205, USA.
| |
Collapse
|
4
|
Youngblood B, Medina JC, Gehlert DR, Schwartz N. EPD1504: a novel μ-opioid receptor partial agonist attenuates obsessive-compulsive disorder (OCD)-like behaviors. Front Psychiatry 2023; 14:1170541. [PMID: 37457777 PMCID: PMC10349350 DOI: 10.3389/fpsyt.2023.1170541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/20/2023] [Indexed: 07/18/2023] Open
Abstract
Low doses of μ-opioid receptor (MOR) agonists rapidly ameliorate symptoms in treatment-resistant obsessive-compulsive disorder (OCD) patients (10-50% of OCD patients). However, the utility of MOR agonists is limited by their safety liabilities. We developed a novel MOR partial agonist (EPD1540) that has an improved respiratory safety profile when compared to buprenorphine. Buprenorphine is a MOR partial agonist primarily used in the treatment of opiate-use disorder, which in investigator-led trials, has been shown to rapidly ameliorate symptoms in treatment-resistant OCD patients. In this study, we show that doses of EPD1504 and buprenorphine that occupy small fractions of MORs in the CNS (approximately 20%) are as effective as fluoxetine at ameliorating OCD-like behaviors in two different rat models (an operant probabilistic reversal task and marble burying). Importantly, effective doses of EPD1504 did not impair either locomotor activity, or respiration under normoxic or hypercapnic conditions. Additionally, EPD1504 had effects comparable to buprenorphine in the conditioned place preference assay. These results indicate that EPD1504 may provide a safer alternative to buprenorphine for the treatment of OCD patients.
Collapse
|
5
|
Tyler BM, Guarnieri M. Long-Acting Opioid Analgesics for Acute Pain: Pharmacokinetic Evidence Reviewed. Vet Sci 2023; 10:372. [PMID: 37368758 DOI: 10.3390/vetsci10060372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Long-acting injectable (LAI) opioid formulations mitigate the harm profiles and management challenges associated with providing effective analgesia for animals. A single dose of a long-acting opioid analgesic can provide up to 72 h of clinically relevant pain management. Yet, few of these new drugs have been translated to products for veterinary clinics. Regulatory pathways allow accelerated drug approvals for generic and biosimilar drugs. These pathways depend on rigorous evidence for drug safety and pharmacokinetic evidence demonstrating bioequivalence between the new and the legacy drug. This report reviews the animal PK data associated with lipid and polymer-bound buprenorphine LAI formulations. Buprenorphine is a widely used veterinary opioid analgesic. Because of its safety profile and regulatory status, buprenorphine is more accessible than morphine, methadone, and fentanyl. This review of PK studies coupled with the well-established safety profile of buprenorphine suggests that the accelerated approval pathways may be available for this new family of LAI veterinary pharmaceuticals.
Collapse
Affiliation(s)
- Betty M Tyler
- Department of Neurosurgery, Johns Hopkins University, 1550 Orleans Street, 1550 Orleans St. CRB II, Baltimore, MD 21287, USA
| | - Michael Guarnieri
- Department of Neurosurgery, Johns Hopkins University, 1550 Orleans Street, 1550 Orleans St. CRB II, Baltimore, MD 21287, USA
| |
Collapse
|
6
|
Janganati V, Salazar P, Parks BJ, Gorman GS, Prather PL, Peterson EC, Alund AW, Moran JH, Crooks PA, Brents LK. Deuterated buprenorphine retains pharmacodynamic properties of buprenorphine and resists metabolism to the active metabolite norbuprenorphine in rats. Front Pharmacol 2023; 14:1123261. [PMID: 37229250 PMCID: PMC10204800 DOI: 10.3389/fphar.2023.1123261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: An active metabolite of buprenorphine (BUP), called norbuprenorphine (NorBUP), is implicated in neonatal opioid withdrawal syndrome when BUP is taken during pregnancy. Therefore, reducing or eliminating metabolism of BUP to NorBUP is a novel strategy that will likely lower total fetal exposure to opioids and thus improve offspring outcomes. Precision deuteration alters pharmacokinetics of drugs without altering pharmacodynamics. Here, we report the synthesis and testing of deuterated buprenorphine (BUP-D2). Methods: We determined opioid receptor affinities of BUP-D2 relative to BUP with radioligand competition receptor binding assays, and the potency and efficacy of BUP-D2 relative to BUP to activate G-proteins via opioid receptors with [35S]GTPγS binding assays in homogenates containing the human mu, delta, or kappa opioid receptors. The antinociceptive effects of BUP-D2 and BUP were compared using the warm-water tail withdrawal assay in rats. Blood concentration versus time profiles of BUP, BUP-D2, and NorBUP were measured in rats following intravenous BUP-D2 or BUP injection. Results: The synthesis provided a 48% yield and the product was ≥99% deuterated. Like BUP, BUP-D2 had sub-nanomolar affinity for opioid receptors. BUP-D2 also activated opioid receptors and induced antinociception with equal potency and efficacy as BUP. The maximum concentration and the area under the curve of NorBUP in the blood of rats that received BUP-D2 were over 19- and 10-fold lower, respectively, than in rats that received BUP. Discussion: These results indicate that BUP-D2 retains key pharmacodynamic properties of BUP and resists metabolism to NorBUP and therefore holds promise as an alternative to BUP.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Paloma Salazar
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Brian J. Parks
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gregory S. Gorman
- Pharmaceutical Sciences Research Institute, McWhorter School of Pharmacy, Samford University, Birmingham, AL, United States
| | - Paul L. Prather
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Eric C. Peterson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Jeffery H. Moran
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- PinPoint Testing, LLC., Little Rock, AR, United States
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lisa K. Brents
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
7
|
Quach T, Hu L, Han S, Lim SF, Senyschyn D, Yadav P, Trevaskis NL, Simpson JS, Porter CJH. Triglyceride-Mimetic Prodrugs of Buprenorphine Enhance Oral Bioavailability via Promotion of Lymphatic Transport. Front Pharmacol 2022; 13:879660. [PMID: 35496278 PMCID: PMC9039622 DOI: 10.3389/fphar.2022.879660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/15/2022] [Indexed: 11/24/2022] Open
Abstract
Buprenorphine (BUP) is a potent opioid analgesic that is widely used for severe pain management and opioid replacement therapy. The oral bioavailability of BUP, however, is significantly limited by first-pass metabolism. Previous studies have shown that triglyceride (TG) mimetic prodrugs of the steroid hormone testosterone circumvent first-pass metabolism by directing drug transport through the intestinal lymphatics, bypassing the liver. The current study expanded this prodrug strategy to BUP. Here different self-immolative (SI) linkers were evaluated to conjugate BUP to the 2 position of the TG backbone via the phenol group on BUP. The SI linkers were designed to promote drug release in plasma. Lipolysis of the prodrug in the intestinal tract was examined via incubation with simulated intestinal fluid (SIF), and potential for parent drug liberation in the systemic circulation was evaluated via incubation in rat plasma. Lymphatic transport and bioavailability studies were subsequently conducted in mesenteric lymph duct or carotid artery-cannulated rats, respectively. TG prodrug derivatives were efficiently transported into the lymphatics (up to 45% of the dose in anaesthetised rats, vs. less than 0.1% for BUP). Incorporation of the SI linkers facilitated BUP release from the prodrugs in the plasma and in concert with high lymphatic transport led to a marked enhancement in oral bioavailability (up to 22-fold) compared to BUP alone. These data suggest the potential to develop an orally bioavailable BUP product which may have advantages with respect to patient preference when compared to current sublingual, transdermal patch or parenteral formulations.
Collapse
Affiliation(s)
- Tim Quach
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- *Correspondence: Sifei Han, ; Christopher J. H. Porter,
| | - Shea F. Lim
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Danielle Senyschyn
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Preeti Yadav
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Natalie L. Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jamie S. Simpson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Christopher J. H. Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- *Correspondence: Sifei Han, ; Christopher J. H. Porter,
| |
Collapse
|
8
|
Schreiner V, Detampel P, Jirkof P, Puchkov M, Huwyler J. Buprenorphine loaded PLGA microparticles: Characterization of a sustained-release formulation. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Xhakaza SP, Khoza LJ, Haripershad AM, Ghazi T, Dhani S, Mutsimhu C, Molopa MJ, Madurai NP, Madurai L, Singh SD, Gopal ND, Kruger HG, Govender T, Chuturgoon A, Naicker T, Baijnath S. Alterations in neurotransmitter levels and transcription factor expression following intranasal buprenorphine administration. Biomed Pharmacother 2021; 138:111515. [PMID: 33752062 DOI: 10.1016/j.biopha.2021.111515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 11/28/2022] Open
Abstract
Buprenorphine is an opioid drug used in the management of pain and the treatment opioid addiction. Like other opioids, it is believed that it achieves these effects by altering functional neurotransmitter pathways and the expression of important transcription factors; cyclic AMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in the brain. However, there is a lack of scientific evidence to support these theories. This study investigated the pharmacodynamic effects of BUP administration by assessing neurotransmitter and molecular changes in the healthy rodent brain. Sprague-Dawley rats (150-200 g) were intranasally administered buprenorphine (0.3 mg/mL) and sacrificed at different time points: 0.25, 0.5, 1, 2, 4, 6, 8 and 24 h post drug administration. LC-MS was used to quantify BUP and neurotransmitters (GABA, GLUT, DA, NE and 5-HT) in the brain, while CREB and BDNF gene expression was determined using qPCR. Results showed that BUP reached a Cmax of 1.21 ± 0.0523 ng/mL after 2 h, with all neurotransmitters showing an increase in their concentration over time, with GABA, GLUT and NE reaching their maximum concentration after 8 h. DA and 5-HT reached their maximum concentrations at 1 h and 24 h, respectively post drug administration. Treatment with BUP resulted in significant upregulation in BDNF expression throughout the treatment period while CREB showed patterns of significant upregulation at 2 and 8 h, and downregulation at 1 and 6 h. This study contributes to the understanding of the pharmacodynamic effects of BUP in opioid addiction by proving that the drug significantly influences NT pathways that are implicated in opioid addiction.
Collapse
Affiliation(s)
- Sanelisiwe P Xhakaza
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Leon J Khoza
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Advaitaa M Haripershad
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Shanel Dhani
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Cosmas Mutsimhu
- Viro Care (SA) Pty Ltd, 12 The Avenue East, Prospecton, Durban 4113, South Africa
| | - Molopa J Molopa
- Viro Care (SA) Pty Ltd, 12 The Avenue East, Prospecton, Durban 4113, South Africa
| | - Nithia P Madurai
- Viro Care (SA) Pty Ltd, 12 The Avenue East, Prospecton, Durban 4113, South Africa
| | - Lorna Madurai
- Viro Care (SA) Pty Ltd, 12 The Avenue East, Prospecton, Durban 4113, South Africa
| | - Sanil D Singh
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Nirmala D Gopal
- Department of Criminology, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Hendrik G Kruger
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Thavendran Govender
- Department of Chemistry, University of Zululand, Private Bag X1001, KwaDlangezwa 3886, South Africa
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Tricia Naicker
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Sooraj Baijnath
- Catalysis and Peptide Research Unit, Department of Pharmaceutical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, 4001, South Africa.
| |
Collapse
|
10
|
Schreiner V, Durst M, Arras M, Detampel P, Jirkof P, Huwyler J. Design and in vivo evaluation of a microparticulate depot formulation of buprenorphine for veterinary use. Sci Rep 2020; 10:17295. [PMID: 33057103 PMCID: PMC7560740 DOI: 10.1038/s41598-020-74230-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/28/2020] [Indexed: 11/09/2022] Open
Abstract
Buprenorphine is a frequently used analgetic agent in veterinary medicine. A major drawback, however, is the short duration of action requiring several daily administrations. We therefore designed a poly-lactic-co-glycolic acid (PLGA) based microparticulate drug formulation for sustained parenteral drug release. Particles were designed to allow for a fast onset of action and a duration of the analgesic effect of at least two days in laboratory mice. Microparticles were produced using a solvent evaporation technique. Release rate was dependent on polymer type and particle size. Spherical particles used for subsequent animal studies had a mean size of 50 µm and contained 4.5% of buprenorphine. Drug release was characterized by an initial burst release of 30% followed by complete release over seven days. In vivo pharmacokinetic experiments in female C57BL/6 J mice confirmed prolonged exposure in plasma and brain tissue and correlated with the pharmacological effect in the hot plate assay or after minor abdominal surgery. No adverse side effects with respect to food and water intake, body weight, local tolerability, or nesting behavior were observed. Our formulation is an attractive alternative to established immediate release formulations. A use for prolonged pain management in laboratory animals is proposed.
Collapse
Affiliation(s)
- Viktoria Schreiner
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Mattea Durst
- Center for Surgical Research, University Hospital Zurich, University Zurich, Zurich, Switzerland
| | - Margarete Arras
- Center for Surgical Research, University Hospital Zurich, University Zurich, Zurich, Switzerland
| | - Pascal Detampel
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Paulin Jirkof
- Center for Surgical Research, University Hospital Zurich, University Zurich, Zurich, Switzerland. .,Department of Animal Welfare and 3Rs, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
11
|
Hammerslag LR, Hofford RS, Kang Q, Kryscio RJ, Beckmann JS, Bardo MT. Changes in fentanyl demand following naltrexone, morphine, and buprenorphine in male rats. Drug Alcohol Depend 2020; 207:107804. [PMID: 31862556 PMCID: PMC6981050 DOI: 10.1016/j.drugalcdep.2019.107804] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND Individuals with opioid use disorder (OUD) exhibit high levels of economic demand for opioids, with high levels of consumption and relative insensitivity to changes in price. Because the medications used to treat OUD in medication-assisted therapy (MAT) act as antagonists or agonists at μ opioid receptors, they may alter the relationship between price and opioid intake. METHODS This study examined demand for a commonly abused synthetic prescription opioid, fentanyl, in male rats following s.c. pre-treatment with naltrexone (0.1-1.0 mg/kg), morphine (0.3-3.0 mg/kg) or buprenorphine (0.3-3.0 mg/kg). We normalized demand curves to intake at the lowest price and estimated effects on elasticity (sensitivity to changes in price). Rats were first trained to earn fentanyl (5 μg/kg/infusion) on a fixed ratio schedule, then they underwent daily training under a threshold procedure designed to produce within-session demand curve estimates. Rats received 14 threshold sessions before undergoing a series of tests encompassing each drug, at each dose. RESULTS Elasticity was increased by pretreatment with naltrexone, morphine or buprenorphine. Morphine also decreased initial intake, when the price for fentanyl was lowest. In contrast, initial intake was increased by naltrexone (according to an inverted-U shaped curve). The effects of naltrexone did not persist after the test session, but morphine and buprenorphine continued affecting demand elasticity 24 h or 48 h after the test, respectively. CONCLUSIONS These results indicate that fentanyl demand is sensitive to blockade or activation of opioid receptors by the drug classes used for MAT in humans.
Collapse
Affiliation(s)
- Lindsey R. Hammerslag
- Department of Psychology, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Rebecca S. Hofford
- Department of Psychology, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Qiwen Kang
- Department of Statistics, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Richard J. Kryscio
- Department of Statistics, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Joshua S. Beckmann
- Department of Psychology, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| | - Michael T. Bardo
- Department of Psychology, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA
| |
Collapse
|
12
|
Hassan R, Pike See C, Sreenivasan S, Mansor SM, Müller CP, Hassan Z. Mitragynine Attenuates Morphine Withdrawal Effects in Rats-A Comparison With Methadone and Buprenorphine. Front Psychiatry 2020; 11:411. [PMID: 32457670 PMCID: PMC7221179 DOI: 10.3389/fpsyt.2020.00411] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/22/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Opiate addiction is a major health problem in many countries. A crucial component of the medical treatment is the management of highly aversive opiate withdrawal signs, which may otherwise lead to resumption of drug taking. In a medication-assisted treatment (MAT), methadone and buprenorphine have been implemented as substitution drugs. Despite MAT effectiveness, there are still limitations and side effects of using methadone and buprenorphine. Thus, other alternative therapies with less side effects, overdosing, and co-morbidities are desired. One of the potential pharmacotherapies may involve kratom's major indole alkaloid, mitragynine, since kratom (Mitragyna speciosa Korth.) preparations have been reported to alleviate opiate withdrawal signs in self-treatment in Malaysian opiate addicts. METHODS Based on the morphine withdrawal model, rats were morphine treated with increasing doses from 10 to 50 mg/kg twice daily over a period of 6 days. The treatment was discontinued on day 7 in order to induce a spontaneous morphine abstinence. The withdrawal signs were measured daily after 24 h of the last morphine administration over a period of 28 abstinence days. In rats that developed withdrawal signs, a drug replacement treatment was given using mitragynine, methadone, or buprenorphine and the global withdrawal score was evaluated. RESULTS The morphine withdrawal model induced profound withdrawal signs for 16 days. Mitragynine (5-30 mg/kg; i.p.) was able to attenuate acute withdrawal signs in morphine dependent rats. On the other hand, smaller doses of methadone (0.5-2 mg/kg; i.p.) and buprenorphine (0.4-1.6 mg/kg; i.p.) were necessary to mitigate these effects. CONCLUSIONS These data suggest that mitragynine may be a potential drug candidate for opiate withdrawal treatment.
Collapse
Affiliation(s)
- Rahimah Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| | - Cheah Pike See
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | - Sasidharan Sreenivasan
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden, Malaysia
| | - Sharif M Mansor
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia
| | - Christian P Müller
- Section of Addiction Medicine, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Minden, Malaysia.,Addiction Behaviour and Neuroplasticity Laboratory, National Neuroscience Institute, Singapore, Singapore
| |
Collapse
|
13
|
Cohier C, Salle S, Fontova A, Mégarbane B, Roussel O. Determination of buprenorphine, naloxone and phase I and phase II metabolites in rat whole blood by LC-MS/MS. J Pharm Biomed Anal 2019; 180:113042. [PMID: 31865207 DOI: 10.1016/j.jpba.2019.113042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/09/2019] [Accepted: 12/09/2019] [Indexed: 11/19/2022]
Abstract
Buprenorphine and buprenorphine/naloxone combination are maintenance treatments used worldwide. However, since their marketing, despite ceiling respiratory effects, poisonings and fatalities have been attributed to buprenorphine misuse and overdose. Therefore, to better understand the mechanisms of buprenorphine-related toxicity in vivo, experimental investigations have been conducted, mainly in the rat. We developed a liquid chromatographic-tandem mass spectrometric (LC-MS/MS) method with electrospray ionization for the simultaneous quantification of buprenorphine, naloxone and their metabolites (norbuprenorphine, buprenorphine glucuronide, norbuprenorphine glucuronide and naloxone glucuronide) in rat whole blood. Compounds were extracted from whole blood by protein precipitation and chromatographically separated using gradient elution of aqueous ammonium formate and methanol in a Raptor Biphenyl core-shell column (100 mm x 3,0 mm x 2,7 μm). Following electrospray ionization, quantification was carried out in the multiple reaction monitoring (MRM) mode by the tandem mass spectrometer API 3200 system. The LC-MS/MS method was validated according to the currently accepted criteria for bioanalytical method validation. The method required small sample volumes (50 μL) and was sensitive with limits of quantification of 6.9, 6.2, 3.6, 3.3, 1.3 and 57.7 ng/mL for buprenorphine, norbuprenorphine, buprenorphine glucuronide, norbuprenorphine glucuronide, naloxone and naloxone glucuronide respectively. The upper limit of quantification was 4000 ng/ml for all the studied compounds. Trueness (88-115 %), repeatability and intermediate precision (both <15%) were in accordance with the international recommendations. The procedure was successfully used to quantify these compounds in the whole blood sample from one rat 24 h after the intravenous administration of buprenorphine/naloxone (30.0/7.5 mg/kg).
Collapse
Affiliation(s)
- Camille Cohier
- Inserm, U1144, Paris, France; Paris-Descartes University, UMR-S 1144, Paris, France; Paris-Diderot University, UMR-S 1144, Paris, France; Forensic Toxicology Unit, Forensic Sciences Institute of the French Gendarmerie, Pontoise, France
| | - Sophie Salle
- Forensic Toxicology Unit, Forensic Sciences Institute of the French Gendarmerie, Pontoise, France
| | - Anne Fontova
- Forensic Toxicology Unit, Forensic Sciences Institute of the French Gendarmerie, Pontoise, France
| | - Bruno Mégarbane
- Inserm, U1144, Paris, France; Paris-Descartes University, UMR-S 1144, Paris, France; Paris-Diderot University, UMR-S 1144, Paris, France; Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Paris, France.
| | - Olivier Roussel
- Inserm, U1144, Paris, France; Paris-Descartes University, UMR-S 1144, Paris, France; Paris-Diderot University, UMR-S 1144, Paris, France; Forensic Toxicology Unit, Forensic Sciences Institute of the French Gendarmerie, Pontoise, France
| |
Collapse
|
14
|
Farkhondeh T, Ashrafizadeh M, Mehrpour O, Roshanravan B, Samarghandian S. Low toxicity in hematological and biomedical parameters caused by bupernorphine in lactating female rats and their newborns. TOXIN REV 2019. [DOI: 10.1080/15569543.2019.1681002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Innovative Medical Research Center, Islamic Azad University, Mashhad, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Omid Mehrpour
- Rocky Mountain Poison and Drug Safety, Denver Health, Denver, CO, USA
| | - Babak Roshanravan
- Medical Student, Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
15
|
Griffin BA, Caperton CO, Russell LN, Cabanlong CV, Wilson CD, Urquhart KR, Martins BS, Zita MD, Patton AL, Alund AW, Owens SM, Fantegrossi WE, Moran JH, Brents LK. In Utero Exposure to Norbuprenorphine, a Major Metabolite of Buprenorphine, Induces Fetal Opioid Dependence and Leads to Neonatal Opioid Withdrawal Syndrome. J Pharmacol Exp Ther 2019; 370:9-17. [PMID: 31028107 DOI: 10.1124/jpet.118.254219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/25/2019] [Indexed: 11/22/2022] Open
Abstract
Buprenorphine is the preferred treatment of opioid use disorder during pregnancy but can cause fetal opioid dependence and neonatal opioid withdrawal syndrome (NOWS). Notably, withdrawal severity is independent of maternal buprenorphine dose, suggesting that interindividual variance in pharmacokinetics may influence risk and severity of NOWS. Using a rat model of NOWS, we tested the hypothesis that clinically relevant doses of the active metabolite norbuprenorphine (NorBUP) can induce in utero opioid dependence, manifested as naltrexone-precipitated withdrawal signs in the neonate. Pregnant Long-Evans rats were implanted with 14-day osmotic minipumps containing vehicle, morphine (positive control), or NorBUP (0.3-10 mg/kg per day) on gestation day 9. By 12 hours post-delivery, an intraperitoneal injection of the opioid antagonist naltrexone (1 or 10 mg/kg) or saline was administered to pups. Precipitated withdrawal signs were graded by raters blinded to treatment conditions. In a separate group, NorBUP concentrations in maternal and fetal blood and brain on gestation day 20 were determined by liquid chromatography-tandem mass spectrometry. Steady-state maternal blood concentrations of NorBUP in dams infused with 1 or 3 mg/kg per day were comparable to values reported in pregnant humans treated with buprenorphine (1.0 and 9.6 ng/ml, respectively), suggesting a clinically relevant dosing regimen. At these doses, NorBUP increased withdrawal severity in the neonate as shown by an evaluation of 10 withdrawal indicators. These findings support the possibility that NorBUP contributes to fetal opioid dependence and NOWS following maternal buprenorphine treatment during pregnancy.
Collapse
Affiliation(s)
- Bryce A Griffin
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Caitlin O Caperton
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Lauren N Russell
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Christian V Cabanlong
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Catheryn D Wilson
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Kyle R Urquhart
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Bradford S Martins
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Marcelle Dina Zita
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Amy L Patton
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Alexander W Alund
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - S Michael Owens
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - William E Fantegrossi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Jeffery H Moran
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| | - Lisa K Brents
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, Arkansas (B.A.G.); Departments of Pharmacology and Toxicology (C.O.C., L.N.R., C.V.C., C.D.W., K.R.U., S.M.O., W.E.F., J.H.M., L.K.B.), Psychiatry (B.S.M.), and Microbiology and Immunology (M.D.Z.), University of Arkansas for Medical Sciences, Little Rock, Arkansas; and PinPoint Testing, LLC, Little Rock, Arkansas (A.L.P., A.W.A., J.H.M.)
| |
Collapse
|
16
|
Evaluation of buprenorphine hydrochloride Pluronic(®) gel formulation in male C57BL/6NCrl mice. Lab Anim (NY) 2017; 45:370-9. [PMID: 27654688 DOI: 10.1038/laban.1106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 08/11/2016] [Indexed: 11/08/2022]
Abstract
Providing adequate analgesia while minimizing handling and stress post-surgery can be challenging. Recently, there have been commercial products made available for providing long acting analgesia in rodents. However, we find there are limitations for use in mice due to the viscosity of the product and the small dosing volumes needed. This project evaluated an in-house compounded formulation of buprenorphine easily made in the laboratory using pharmaceutical grade products. The release of buprenorphine was evaluated when compounded with two types of hydrogels (Pluronic(®) F-127 and F-68). Mice given buprenorphine in hydrogel (BP) demonstrated higher serum levels of buprenorphine for a longer period of time compared to mice given standard buprenorphine (Bup). However, the rate of decline in serum levels between the groups was similar; thus, it is more likely that the higher buprenorphine concentration seen in the BP group is due to the higher dose of buprenorphine given, rather than a slower release of product. Feed consumption was decreased in both groups one day after dosing; however, there was no difference in body weights. Increased activity in the open field was observed with both buprenorphine formulations, and lipemia was observed in mice given BP which persisted to at least 96 h. Based on our results, we conclude that this formulation did not sustain the release of buprenorphine or eliminate the increased activity commonly seen in mice given buprenorphine. In addition, the lipemia may confound research parameters, especially in cardiac studies and lipid metabolism studies. Therefore, we cannot recommend this formulation for use.
Collapse
|
17
|
Barbarossa A, Rambaldi J, Giunti M, Zaghini A, Cunto M, Zambelli D, Valgimigli S, Santoro F, Romagnoli N. Pharmacokinetics of buprenorphine following constant rate infusion for postoperative analgesia in dogs undergoing ovariectomy. Vet Anaesth Analg 2017; 44:435-443. [DOI: 10.1016/j.vaa.2016.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/16/2016] [Accepted: 05/03/2016] [Indexed: 10/20/2022]
|
18
|
Hagelberg NM, Fihlman M, Hemmilä T, Backman JT, Laitila J, Neuvonen PJ, Laine K, Olkkola KT, Saari TI. Rifampicin decreases exposure to sublingual buprenorphine in healthy subjects. Pharmacol Res Perspect 2016; 4:e00271. [PMID: 28097004 PMCID: PMC5226287 DOI: 10.1002/prp2.271] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 09/30/2016] [Indexed: 01/03/2023] Open
Abstract
Buprenorphine is mainly metabolized by the cytochrome P450 (CYP) 3A4 enzyme. The aim of this study was to evaluate the role of first‐pass metabolism in the interaction of rifampicin and analgesic doses of buprenorphine. A four‐session paired cross‐over study design was used. Twelve subjects ingested either 600 mg oral rifampicin or placebo once daily in a randomized order for 7 days. In the first part of the study, subjects were given 0.6‐mg (placebo phase) or 0.8‐mg (rifampicin phase) buprenorphine sublingually on day 7. In the second part of the study, subjects received 0.4‐mg buprenorphine intravenously. Plasma concentrations of buprenorphine and urine concentrations of buprenorphine and its primary metabolite norbuprenorphine were measured over 18 h. Adverse effects were recorded. Rifampicin decreased the mean area under the dose‐corrected plasma concentration–time curve (AUC0–18) of sublingual buprenorphine by 25% (geometric mean ratio (GMR): 0.75; 90% confidence interval (CI) of GMR: 0.60, 0.93) and tended to decrease the bioavailability of sublingual buprenorphine, from 22% to 16% (P = 0.31). Plasma concentrations of intravenously administered buprenorphine were not influenced by rifampicin. The amount of norbuprenorphine excreted in the urine was decreased by 65% (P < 0.001) and 52% (P < 0.001) after sublingual and intravenous administration, respectively, by rifampicin. Adverse effects were frequent. Rifampicin decreases the exposure to sublingual but not intravenous buprenorphine. This can be mainly explained by an enhancement of CYP3A‐mediated first‐pass metabolism, which sublingual buprenorphine only partially bypasses. Concomitant use of rifampicin and low‐dose sublingual buprenorphine may compromise the analgesic effect of buprenorphine.
Collapse
Affiliation(s)
- Nora M Hagelberg
- Department of Anaesthesiology, Intensive Care and Pain Medicine University of Turku and Turku University Hospital Turku Finland
| | - Mari Fihlman
- Department of Anaesthesiology, Intensive Care and Pain Medicine University of Turku and Turku University Hospital Turku Finland
| | - Tuija Hemmilä
- Department of Anaesthesiology, Intensive Care and Pain Medicine University of Turku and Turku University Hospital Turku Finland
| | - Janne T Backman
- Department of Clinical Pharmacology University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Jouko Laitila
- Department of Clinical Pharmacology University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Kari Laine
- Department of Pharmacology Drug Development and Therapeutics University of Turku Turku Finland; Medbase Ltd Turku Finland
| | - Klaus T Olkkola
- Department of Anaesthesiology, Intensive Care and Pain Medicine University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Teijo I Saari
- Department of Anaesthesiology, Intensive Care and Pain Medicine University of Turku and Turku University Hospital Turku Finland
| |
Collapse
|
19
|
Joshi A, Halquist M, Konsoula Z, Liu Y, Jones JP, Heidbreder C, Gerk PM. Improving the oral bioavailability of buprenorphine: an in-vivo proof of concept. J Pharm Pharmacol 2016; 69:23-31. [DOI: 10.1111/jphp.12652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/18/2016] [Indexed: 12/20/2022]
Abstract
Abstract
Objectives
The aim of this study was to improve the oral bioavailability of buprenorphine by inhibiting presystemic metabolism via the oral co-administration of ‘Generally Recognized as Safe’ compounds, thus providing an orally administered drug product with less variability and comparable or higher exposure compared with the sublingual route.
Methods
The present studies were performed in Sprague Dawley rats following either intravenous or oral administration of buprenorphine/naloxone and oral co-administration of ‘Generally Recognized as Safe’ compounds referred to as ‘adjuvants’. Plasma samples were collected up to 22 h postdosing followed by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) analysis.
Key findings
The adjuvants increased Cmax (21 ± 16 ng/ml vs 75 ± 33 ng/ml; 3.6-fold) and AUC(0–22 h) (10.6 ± 8.11 μg min/ml vs 22.9 ± 11.7 μg min/ml; 2.2-fold) values of buprenorphine (control vs adjuvant-treated, respectively). The absolute oral bioavailability of buprenorphine doubled (from 1.24% to 2.68%) in the presence of the adjuvants.
Conclusions
One may suggest that the adjuvant treatment most likely inhibited the presystemic metabolic enzymes, thus decreasing the intestinal ‘first-pass effect’ on buprenorphine. Additional studies are now required to further explore the concept of inhibiting presystemic metabolism of buprenorphine by adjuvants to potentially increase the oral bioavailability of buprenorphine.
Collapse
Affiliation(s)
- Anand Joshi
- Department of Pharmaceutics, VCU School of Pharmacy, Richmond, VA, USA
| | - Matthew Halquist
- Department of Pharmaceutics, VCU School of Pharmacy, Richmond, VA, USA
| | | | - Yongzhen Liu
- Global Research & Development, Indivior Inc., Richmond, VA, USA
| | - J P Jones
- Global Research & Development, Indivior Inc., Richmond, VA, USA
| | | | - Phillip M Gerk
- Department of Pharmaceutics, VCU School of Pharmacy, Richmond, VA, USA
| |
Collapse
|
20
|
Joshi A, Parris B, Liu Y, Heidbreder C, Gerk PM, Halquist M. Quantitative determination of buprenorphine, naloxone and their metabolites in rat plasma using hydrophilic interaction liquid chromatography coupled with tandem mass spectrometry. Biomed Chromatogr 2016; 31. [DOI: 10.1002/bmc.3785] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/23/2016] [Accepted: 06/30/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Anand Joshi
- Department of Pharmaceutics; VCU School of Pharmacy; Richmond VA 23298 USA
| | - Brian Parris
- Department of Pharmaceutics; VCU School of Pharmacy; Richmond VA 23298 USA
| | - Yongzhen Liu
- Global Research & Development; Indivior Inc.; Richmond VA 23235 USA
| | | | - Phillip M. Gerk
- Department of Pharmaceutics; VCU School of Pharmacy; Richmond VA 23298 USA
| | - Matthew Halquist
- Department of Pharmaceutics; VCU School of Pharmacy; Richmond VA 23298 USA
| |
Collapse
|
21
|
Gustavsen KA, Guzman DSM, Knych HK, Petritz OA, Olsen GH, Paul-Murphy JR. Pharmacokinetics of buprenorphine hydrochloride following intramuscular and intravenous administration to American kestrels (Falco sparverius). Am J Vet Res 2014; 75:711-5. [PMID: 25061701 DOI: 10.2460/ajvr.75.8.711] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine the pharmacokinetics of buprenorphine hydrochloride after IM and IV administration to American kestrels (Falco sparverius). ANIMALS 13 healthy 3-year-old captive-bred American kestrels. PROCEDURES Buprenorphine hydrochloride (0.6 mg/kg) was administered IM to all birds. Blood samples were collected at 9 times, ranging from 5 minutes to 9 hours after drug administration. Plasma buprenorphine concentrations were measured by use of tandem liquid chromatography-mass spectrometry. Pharmacokinetic parameters were determined by use of least squares linear regression and noncompartmental analysis of naïve pooled data. After a washout period of 2 weeks, the same dose of buprenorphine was administered IV to all birds and blood samples were collected at the same times after drug administration. RESULTS Maximum plasma buprenorphine concentration was achieved within 5 minutes after IM administration. For IM administration, bioavailability was 94.8% and elimination half-life was 92.1 minutes. For IV administration, steady-state volume of distribution was 4,023.8 mL/kg, plasma clearance was 49.2 mL/min/kg, and elimination half-life was 105.5 minutes. CONCLUSIONS AND CLINICAL RELEVANCE Buprenorphine was rapidly absorbed, and bioavailability was good after IM administration to American kestrels. Plasma buprenorphine concentrations were > 1 ng/mL for 9 hours after both IM and IV administration. These results, in combination with those of a pharmacodynamic study, suggested that the analgesic effects of buprenorphine could last at least 6 to 9 hours in this species. Further investigations of the duration of analgesic effects, multiple-dose protocols, and potential adverse effects of buprenorphine are warranted in American kestrels and other raptors.
Collapse
Affiliation(s)
- Kate A Gustavsen
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California-Davis, Davis CA 95616
| | | | | | | | | | | |
Collapse
|
22
|
Comparison of toxicity associated with nonmedical use of benzodiazepines with buprenorphine or methadone. Drug Alcohol Depend 2014; 138:118-23. [PMID: 24629782 DOI: 10.1016/j.drugalcdep.2014.02.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/05/2014] [Accepted: 02/07/2014] [Indexed: 11/22/2022]
Abstract
BACKGROUND Polysubstance use is prevalent in individuals using buprenorphine or methadone nonmedically, with benzodiazepines being a common co-ingestant. The objective of this study was to compare the severity of buprenorphine and methadone toxicity with concomitant use of benzodiazepines. METHODS A retrospective analysis of buprenorphine and methadone cases from November 1, 2002 to December 31, 2010 reported to the American Association of Poison Control Centers' National Poison Data System (NPDS) was conducted. INCLUSION CRITERIA age ≥ 18 years, nonmedical use of methadone with benzodiazepines (methadone-BZD) or buprenorphine with benzodiazepines (BUP-BZD), and case followed to a documented outcome. Cases with co-ingestants other than benzodiazepines were excluded. Clinical effects, treatments, disposition and final medical outcomes were evaluated. RESULTS There were 692 methadone-BZD cases and 72 BUP-BZD cases. Clinical effects in methadone-BZD and BUP-BZD groups were lethargy (71.1%, 59.7%), respiratory depression (29.0%, 15.3%), coma (22.4%, 5.6%), respiratory arrest (4.5%, 0), hypotension (11.8%, 2.8%) and cardiac arrest (1.9%, 0), respectively. Patients in the methadone-BZD group were four-times more likely to receive naloxone (60.4% vs 15.3%) or be intubated (16.3% vs 4.2%) than in the BUP-BZD group. Hospitalization rates were highest for methadone-BZD patients with 67.3% receiving medical admissions compared to 43.3% of BUP-BZD patients. Outcomes were more serious for methadone-BZD cases (p<0.0001); while there were no BUP-BZD deaths, exposure to methadone-BZD yielded 16 deaths. CONCLUSIONS Nonmedical use of benzodiazepines with methadone is associated with higher hospitalization rates, greater ICU utilization rates and considerably worse medical outcomes when compared to nonmedical use of benzodiazepines with buprenorphine.
Collapse
|
23
|
Guarnieri M, Brayton C, DeTolla L, Forbes-McBean N, Sarabia-Estrada R, Zadnik P. Safety and efficacy of buprenorphine for analgesia in laboratory mice and rats. Lab Anim (NY) 2013; 41:337-43. [PMID: 23079917 DOI: 10.1038/laban.152] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 06/08/2012] [Indexed: 11/09/2022]
Abstract
Buprenorphine is a long-acting opiate with a high therapeutic index. The authors review the pharmacology, toxicity, analgesic effects and delivery of buprenorphine for use in laboratory mice and rats. Buprenorphine-based analgesic therapy has a substantial record of safety, and there is growing evidence of its effectiveness for treating post-operative pain. Nonetheless, more research is needed to determine optimal delivery systems and analgesic regimens for pain therapy in laboratory animals.
Collapse
|
24
|
Becerra L, Upadhyay J, Chang PC, Bishop J, Anderson J, Baumgartner R, Schwarz AJ, Coimbra A, Wallin D, Nutile L, George E, Maier G, Sunkaraneni S, Iyengar S, Evelhoch JL, Bleakman D, Hargreaves R, Borsook D. Parallel buprenorphine phMRI responses in conscious rodents and healthy human subjects. J Pharmacol Exp Ther 2013; 345:41-51. [PMID: 23370795 DOI: 10.1124/jpet.112.201145] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pharmacological magnetic resonance imaging (phMRI) is one method by which a drug's pharmacodynamic effects in the brain can be assessed. Although phMRI has been frequently used in preclinical and clinical settings, the extent to which a phMRI signature for a compound translates between rodents and humans has not been systematically examined. In the current investigation, we aimed to build on recent clinical work in which the functional response to 0.1 and 0.2 mg/70 kg i.v. buprenorphine (partial µ-opioid receptor agonist) was measured in healthy humans. Here, we measured the phMRI response to 0.04 and 0.1 mg/kg i.v. buprenorphine in conscious, naive rats to establish the parallelism of the phMRI signature of buprenorphine across species. PhMRI of 0.04 and 0.1 mg/kg i.v. buprenorphine yielded dose-dependent activation in a brain network composed of the somatosensory cortex, cingulate, insula, striatum, thalamus, periaqueductal gray, and cerebellum. Similar dose-dependent phMRI activation was observed in the human phMRI studies. These observations indicate an overall preservation of pharmacodynamic responses to buprenorphine between conscious, naive rodents and healthy human subjects, particularly in brain regions implicated in pain and analgesia. This investigation further demonstrates the usefulness of phMRI as a translational tool in neuroscience research that can provide mechanistic insight and guide dose selection in drug development.
Collapse
Affiliation(s)
- Lino Becerra
- Imaging Consortium for Drug Development, P.A.I.N. Group, Harvard Medical School, Children’s Hospital of Boston, Waltham, Massachusetts 02453, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Targeting tacrolimus to deeper layers of skin with improved safety for treatment of atopic dermatitis-Part II: in vivo assessment of dermatopharmacokinetics, biodistribution and efficacy. Int J Pharm 2012; 434:70-9. [PMID: 22609427 DOI: 10.1016/j.ijpharm.2012.04.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/12/2012] [Accepted: 04/21/2012] [Indexed: 01/22/2023]
Abstract
The objective of present investigation was to study in vivo behavior of tacrolimus-loaded lipid-nanoparticles (T-LN) to understand its targeting potential for treatment of atopic-dermatitis-(AD). T-LN have shown significantly improved drug penetration to deeper epidermal and dermal skin-layers than commercial ointment-Protopic(®) and effectively reached target dendritic-immune-cells, responsible for immunopathogenesis of AD. Due to enhanced penetrability of T-LN, it became necessary to evaluate the toxicity of the nanocarrier and the drug at non-target tissues. This paper evaluates dermatopharmacokinetics (DPK), biodistribution, efficacy and safety of T-LN in comparison to Protopic(®) as reference. In vivo DPK in guinea pigs showed 3.02-fold higher bioavailability while γ-scintigraphy in albino-rats demonstrated 1.5-fold rapid penetration of radioactivity in skin for T-LN. Biodistribution in albino-rats revealed restricted localization at the target-skin-area with no general spreading to other body organs suggesting targeting potential of T-LN. In vivo efficacy studies in BALB/c mice showed highly efficient suppression of inflammatory AD-like skin-lesions with T-LN than reference and placebo. Dermal toxicity-studies revealed keratosis and collagenous mass-infiltration with repeated application of reference however interestingly, T-LN treated group showed no evident toxicity demonstrating significantly improved safety. Thus T-LN offered improved penetration to the target site without any toxic-effects and would represent an efficient and commercially viable alternative for AD treatment.
Collapse
|
26
|
Abstract
BACKGROUND The metabolism of opioids is critical to consider for multiple reasons. The most commonly prescribed opioid agents often have metabolites that are active and are the source of both analgesic activity and an increased incidence of adverse events. Many opioids are metabolized by cytochrome P450 enzymes. Polymorphisms in cytochrome P450 genes and inhibition or induction of cytochrome P450 enzymes by coadministered drugs may significantly impact the systemic concentration of opioids and their metabolites and the associated efficacy or adverse events. METHODS This is a narrative review of the metabolism of various opioids that will highlight the impact of their active metabolites, and the potential impact of cytochrome P450 activity on analgesic activity. RESULTS An understanding of "opioid metabolic machinery," cytochrome P450 activity, and drug-drug interactions in the context of opioid selection may benefit clinicians and patients alike. CONCLUSIONS A greater appreciation of the metabolism of commonly prescribed opioid analgesics and the impact of their active metabolites on efficacy and safety may aid prescribers in tailoring care for optimal outcomes.
Collapse
|
27
|
Kukanich B. Clinical interpretation of pharmacokinetic and pharmacodynamic data in zoologic companion animal species. Vet Clin North Am Exot Anim Pract 2010; 14:1-20. [PMID: 21074699 DOI: 10.1016/j.cvex.2010.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The treatment and prevention of pain in zoologic companion animals is difficult because of the lack of data available on the safety and efficacy of analgesics. Pharmacokinetic (PK)-pharmacodynamic (PD) studies integrate changes in drug concentrations and changes in the drug's effect. All experimental studies assessing the PDs of analgesics have limitations in animals, but the data provided by experimental studies are valuable in designing dosages. Placebo-controlled, randomized, and blinded clinical trials provide the best PK and PD data, but are rarely performed in major veterinary species because of the number of animals required for the study, lack of preliminary PK and PD data in a given species, species-specific differences in PK and PD, and ethical and toxicologic concerns. The usefulness and limitations as well as considerations for interpreting PK, PD, and controlled clinical studies are discussed. An example of allometric analysis of buprenorphine in mammals is also included.
Collapse
Affiliation(s)
- Butch Kukanich
- College of Veterinary Medicine, Kansas State University, 228 Coles Hall, Manhattan, KS 66506-5802, USA.
| |
Collapse
|
28
|
Thomas A, Déglon J, Steimer T, Mangin P, Daali Y, Staub C. On-line desorption of dried blood spots coupled to hydrophilic interaction/reversed-phase LC/MS/MS system for the simultaneous analysis of drugs and their polar metabolites. J Sep Sci 2010; 33:873-9. [PMID: 20029845 DOI: 10.1002/jssc.200900593] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
An assay for the simultaneous analysis of pharmaceutical compounds and their metabolites from micro-whole blood samples (i.e. 5 microL) was developed using an on-line dried blood spot (on-line DBS) device coupled with hydrophilic interaction/reversed-phase (HILIC/RP) LC/MS/MS. Filter paper is directly integrated to the LC device using a homemade inox desorption cell. Without any sample pretreatment, analytes are desorbed from the paper towards an automated system of valves linking a zwitterionic-HILIC column to an RP C18 column. In the same run, the polar fraction is separated by the zwitterionic-HILIC column while the non-polar fraction is eluted on the RP C18. Both fractions are detected by IT-MS operating in full scan mode for the survey scan and in product ion mode for the dependant scan using an ESI source. The procedure was evaluated by the simultaneous qualitative analysis of four probes and their relative phase I and II metabolites spiked in whole blood. In addition, the method was successfully applied to the in vivo monitoring of buprenorphine metabolism after the administration of an intraperitoneal injection of 30 mg/kg on adult female Wistar rat.
Collapse
Affiliation(s)
- Aurélien Thomas
- Unit of Toxicology, University Center of Legal Medicine, Geneva, Switzerland.
| | | | | | | | | | | |
Collapse
|
29
|
Andaluz A, Moll X, Abellán R, Ventura R, Carbó M, Fresno L, García F. Pharmacokinetics of buprenorphine after intravenous administration of clinical doses to dogs. Vet J 2009; 181:299-304. [DOI: 10.1016/j.tvjl.2008.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 03/03/2008] [Accepted: 03/04/2008] [Indexed: 10/22/2022]
|
30
|
Cho HJ, Staikopoulos V, Furness JB, Jennings EA. Inflammation-induced increase in hyperpolarization-activated, cyclic nucleotide-gated channel protein in trigeminal ganglion neurons and the effect of buprenorphine. Neuroscience 2009; 162:453-61. [PMID: 19409968 DOI: 10.1016/j.neuroscience.2009.04.063] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 04/17/2009] [Accepted: 04/26/2009] [Indexed: 12/20/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are active at resting membrane potential and thus contribute to neuronal excitability. Their increased activity has recently been demonstrated in models of nerve injury-induced pain. The major aim of the current study was to investigate altered HCN channel protein expression in trigeminal sensory neurons following inflammation of the dura. HCN1 and HCN2 channel immunoreactivity was observed on the membranes of medium- to large-sized trigeminal ganglion neurons with 76% and 85% of HCN1 and HCN2 expressing neurons also containing the 200 kDa neurofilament protein (associated with myelinated fibers). Western immunoblots of lysates from rat trigeminal ganglia also showed bands with appropriate molecular weights for HCN1 and HCN2. Three days after application of complete Freund's adjuvant (CFA) to the dura mater, Western blot band densities were significantly increased; compared to control, to 166% for HCN1 and 284% for HCN2 channel protein. The band densities were normalized against alpha-actin. In addition, the number of retrogradely labeled neurons from the dura expressing HCN1 and HCN2 was significantly increased to 247% (HCN1) and 171% (HCN2), three days after inflammation. When the opioid receptor partial agonist, buprenorphine, was given systemically, immediately after CFA, the inflammation-induced increase in HCN protein expression in both Western blot and immunohistochemical experiments was not observed. These results suggest that HCN1 and HCN2 are involved in inflammation-induced sensory neuron hyperexcitability, and indicate that an opioid receptor agonist can reverse the protein upregulation.
Collapse
Affiliation(s)
- H-J Cho
- Department of Anatomy and Cell Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | |
Collapse
|
31
|
Krotscheck U, Boothe DM, Little AA. Pharmacokinetics of buprenorphine following intravenous administration in dogs. Am J Vet Res 2008; 69:722-7. [DOI: 10.2460/ajvr.69.6.722] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
Wu CH, Yang SC, Wang YS, Chen BG, Lin CC, Liu RH. Evaluation of various derivatization approaches for gas chromatography–mass spectrometry analysis of buprenorphine and norbuprenorphine. J Chromatogr A 2008; 1182:93-112. [DOI: 10.1016/j.chroma.2007.11.063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2007] [Revised: 11/18/2007] [Accepted: 11/22/2007] [Indexed: 01/31/2023]
|
33
|
Bruijnzeel AW, Marcinkiewcz C, Isaac S, Booth MM, Dennis DM, Gold MS. The effects of buprenorphine on fentanyl withdrawal in rats. Psychopharmacology (Berl) 2007; 191:931-41. [PMID: 17211652 DOI: 10.1007/s00213-006-0670-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Accepted: 12/05/2006] [Indexed: 11/30/2022]
Abstract
RATIONALE Fentanyl is a potent mu-opioid receptor agonist that is widely used for the treatment of severe chronic pain. Discontinuation of fentanyl administration has been shown to induce a negative emotional state. OBJECTIVES The aim of the present studies was to investigate the effects of the partial mu-opioid receptor agonist buprenorphine on the negative emotional state associated with precipitated and spontaneous fentanyl withdrawal in rats. MATERIALS AND METHODS Fentanyl and saline were chronically administered via osmotic minipumps. A discrete-trial intracranial self-stimulation procedure was used to provide a measure of brain reward function. Somatic signs were recorded from a checklist of opioid abstinence signs. RESULTS Naloxone induced a deficit in brain reward function in rats chronically treated with fentanyl. Buprenorphine dose-dependently prevented the naloxone-induced deficit in brain reward function. Discontinuation of fentanyl administration was also associated with a deficit in brain reward function. After explantation of the minipumps, the administration of buprenorphine induced a potentiation of brain reward function in the fentanyl-withdrawing rats, but did not affect brain reward function of saline-treated control rats. Buprenorphine prevented the somatic withdrawal signs associated with spontaneous fentanyl withdrawal and attenuated the somatic signs associated with precipitated fentanyl withdrawal. CONCLUSIONS Buprenorphine prevents affective and somatic fentanyl withdrawal signs. Moreover, buprenorphine is rewarding in rats previously exposed to fentanyl, but not in opioid-naïve rats. This pattern of results suggests that buprenorphine may be an effective treatment for the anhedonic-state associated with fentanyl withdrawal, but further study of buprenorphine's abuse potential is warranted.
Collapse
Affiliation(s)
- Adrie W Bruijnzeel
- Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, 100 S. Newell Dr., 100256, Gainesville, FL 32610-0244, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Yassen A, Kan J, Olofsen E, Suidgeest E, Dahan A, Danhof M. Pharmacokinetic-pharmacodynamic modeling of the respiratory depressant effect of norbuprenorphine in rats. J Pharmacol Exp Ther 2007; 321:598-607. [PMID: 17283225 DOI: 10.1124/jpet.106.115972] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The objective of this investigation was to characterize the pharmacokinetic-pharmacodynamic (PK-PD) correlation of buprenorphine's active metabolite norbuprenorphine for the effect on respiration in rats. Following i.v. administration in rats (dose range 0.32-1.848 mg), the time course of the concentration in plasma was determined in conjunction with the effect in ventilation as determined with a novel whole-body plethysmography technique. The PK of norbuprenorphine was best described by a three-compartment PK model with nonlinear elimination. A saturable biophase distribution model with a power PD model described the PK-PD relationship best. No saturation of the effect at high concentrations was observed, indicating that norbuprenorphine acts as a full agonist with regard to respiratory depression. Moreover, analysis of the hysteresis based on the combined receptor association-dissociation biophase distribution model yielded high values of the rate constants for receptor association and dissociation, indicating that these processes are not rate-limiting. In a separate analysis, the time course of the plasma concentrations of buprenorphine and norbuprenorphine following administration of both the parent drug and the metabolite were simultaneously analyzed based on a six-compartment PK model with nonlinear elimination of norbuprenorphine. This analysis showed that following i.v. administration, 10% of the administered dose of buprenorphine is converted into norbuprenorphine. By simulation it is shown that following i.v. administration of buprenorphine, the concentrations of norbuprenorphine reach values that are well below the values causing an effect on respiration.
Collapse
Affiliation(s)
- Ashraf Yassen
- Leiden/Amsterdam Center for Drug Research, Division of Pharmacology, Gorlaeus Laboratories, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
35
|
Mégarbane B, Hreiche R, Pirnay S, Marie N, Baud FJ. Does high-dose buprenorphine cause respiratory depression?: possible mechanisms and therapeutic consequences. ACTA ACUST UNITED AC 2007; 25:79-85. [PMID: 16958555 DOI: 10.2165/00139709-200625020-00002] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Buprenorphine is an opioid agonist-antagonist with a 'ceiling effect' for respiratory depression. Compared with methadone, its unique pharmacology offers practical advantages and enhanced safety when prescribed as recommended and supervised by a physician. Buprenorphine has been approved in several countries as an efficient and safe maintenance therapy for heroin addiction. Its use resulted in a salutary effect with a reduction in heroin overdose-related deaths in countries that implemented office-based buprenorphine maintenance. In France, however, where high-dose buprenorphine has been marketed since 1996, several cases of asphyxic deaths were reported among addicts treated with buprenorphine. Death resulted from buprenorphine intravenous misuse or concomitant sedative drug ingestion, such as benzodiazepines. In these situations of abuse, misuse, or in association with elevated doses of psychotropic drugs, buprenorphine may cause severe respiratory depression. Unlike other opiates, the respiratory effects from buprenorphine are not responsive to naloxone. However, the exact mechanism of buprenorphine-induced effects on ventilation is still unknown. The role of norbuprenorphine, the main N-dealkylated buprenorphine metabolite with potent respiratory depressor activity, also remains unclear. Experimental studies investigating the respiratory effects of combinations of high doses of buprenorphine and benzodiazepines suggested that this drug-drug interaction may result from a pharmacodynamic interaction. A pharmacokinetic interaction between buprenorphine and flunitrazepam is also considered. As there are many questions regarding the possible dangers of death or respiratory depression associated with buprenorphine use, we aimed to present a comprehensive critical review of the published clinical and experimental studies on buprenorphine respiratory effects.
Collapse
Affiliation(s)
- Bruno Mégarbane
- INSERM U705, CNRS, UMR 7157, Université Paris 7, Université Paris 5, Hôpital Fernand Widal, Paris, France.
| | | | | | | | | |
Collapse
|
36
|
Hreiche R, Mégarbane B, Pirnay S, Borron SW, Monier C, Risède P, Milan N, Descatoire V, Pessayre D, Baud FJ. Dexamethasone hepatic induction in rats subsequently treated with high dose buprenorphine does not lead to respiratory depression. Toxicol Appl Pharmacol 2006; 217:352-62. [PMID: 17084876 DOI: 10.1016/j.taap.2006.09.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 09/22/2006] [Accepted: 09/25/2006] [Indexed: 10/24/2022]
Abstract
In humans, asphyxic deaths and severe poisonings have been attributed to high-dosage buprenorphine, a maintenance therapy for heroin addiction. However, in rats, intravenous buprenorphine at doses up to 90 mg kg(-1) was not associated with significant effects on arterial blood gases. In contrast, norbuprenorphine, the buprenorphine major cytochrome P450 (CYP) 3A-derived metabolite, is a potent respiratory depressant. Thus, our aim was to study the consequences of CYP3A induction on buprenorphine-associated effects on resting ventilation in rats. We investigated the effects on ventilation of 30 mg kg(-1) buprenorphine alone or following cytochrome P450 (CYP) 3A induction with dexamethasone, using whole body plethysmography (N=24) and arterial blood gases (N=12). Randomized animals in 4 groups received sequential intraperitoneal dosing with: (dexamethasone [days 1-3]+buprenorphine [day 4]), (dexamethasone solvent [days 1-3]+buprenorphine [day 4]), (dexamethasone [days 1-3]+buprenorphine solvent [day 4]), or (dexamethasone solvent [days 1-3]+buprenorphine solvent [day 4]). Buprenorphine alone caused a significant rapid and sustained increase in the inspiratory time (P<0.001), without significant effects on the respiratory frequency, the tidal volume, the minute volume, or arterial blood gases. In dexamethasone-pretreated rats, there was no significant alteration in the respiratory parameters, despite CYP3A induction and significant increase of the ratio of plasma norbuprenorphine-to-buprenorphine concentrations. In conclusion, dexamethasone did not modify the effects of 30 mg kg(-1) buprenorphine on rat ventilation. Our results suggest a limited role of drug-mediated CYP3A induction in the occurrence of buprenorphine-attributed respiratory depression in addicts.
Collapse
Affiliation(s)
- Raymond Hreiche
- INSERM U705, CNRS UMR 7157, Université Paris 7, Université Paris 5, Hôpital Fernand Widal, 75010 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Englberger W, Kögel B, Friderichs E, Strassburger W, Germann T. Reversibility of opioid receptor occupancy of buprenorphine in vivo. Eur J Pharmacol 2006; 534:95-102. [PMID: 16490191 DOI: 10.1016/j.ejphar.2006.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2005] [Revised: 12/22/2005] [Accepted: 01/10/2006] [Indexed: 11/17/2022]
Abstract
The slow association and incomplete dissociation of buprenorphine from opioid receptors observed in vitro have been suggested to reduce the accessibility of opioid receptors in vivo. If so, it might be expected that buprenorphine continues to occupy opioid receptors long after the antinociceptive activity has dissipated. To examine this hypothesis, buprenorphine (46.4 microg/kg i.v.) was administered to rats 1, 2, 4 or 8 h before isolation of their forebrain membranes and the maximal binding capacity (Bmax) for [3H]-[D-Ala2, N-methyl-Phe4-Gly5-ol]-enkephalin ([3H]DAMGO) was determined to measure the number of mu-opioid receptor binding sites remaining. Extent and duration of the reduction of Bmax by buprenorphine (ED50 11.2 microg/kg 1 h post-application) correlated with the antinociceptive activity in the rat tail flick (ED50 16.4 microg/kg i.v. 1 h post-application). At 8 h after administration there was still residual antinociception but no further attenuation of Bmax was detectable. Thus receptor occupancy by buprenorphine does not cause impairment of mu-opioid receptor accessibility beyond the duration of its antinociceptive activity. Therefore, no impairment of antinociception in the case of an opioid switch is to be expected.
Collapse
Affiliation(s)
- Werner Englberger
- Department of Molecular Pharmacology, Research Center, Grünenthal GmbH, Aachen, Germany
| | | | | | | | | |
Collapse
|
38
|
Quentin T, Debruyne D, Lelong-Boulouard V, Poisnel G, Barre L, Coquerel A. Clorazepate affects cell surface regulation of delta and kappa opioid receptors, thereby altering buprenorphine-induced adaptation in the rat brain. Brain Res 2005; 1063:84-95. [PMID: 16269137 DOI: 10.1016/j.brainres.2005.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 09/01/2005] [Accepted: 09/25/2005] [Indexed: 11/18/2022]
Abstract
Concomitant abuse of buprenorphine (BPN) and benzodiazepines (BZD) may relate to a pharmacodynamic interaction between the two. The objective of the present work was to investigate the acute and chronic effects of clorazepate (CRZ) alone or in combination with BPN on selective kappa opiate tritiated ligand [3H]-U69 593 and delta opiate radioligand [3H]-deltorphine II binding in the rat brain. Bmax (maximal receptor density) and Kd (the dissociation constant) were directly determined at different brain regions of interest (ROI) selected for high densities of kappa and/or delta receptors in rats treated with BPN and/or CRZ. The agents were administered either once or for 21 consecutive days. Differences in Bmax and Kd (for both specific ligands) were related to drug treatment and receptor location. Globally, single BPN administration induced no changes in kappa or delta opiate receptor binding, whereas repeated BPN administration up-regulated kappa receptor density and decreased delta affinity. At the kappa receptor level, repeated administration of CRZ acted only on Kd, whereas the delta receptor was up-regulated. Repeated addition of CRZ to BPN had no effect on kappa receptor Bmax versus chronic controls. By significantly decreasing Bmax, CRZ nullified the effect of chronic BPN on the kappa receptor. The modifications were strongest in the nucleus accumbens, where both types of receptor occur. Treatments had region-selective effects in some brain areas, such as the amygdala, periaqueductal gray matter, hypothalamus and caudate putamen. Increased mu and delta receptor densities would be expected to provide reinforcement by enhancing reward, and impairment of kappa receptor availability would be expected to decrease aversion. The effects described are likely to influence addictive behavior among people abusing BZD and BPN.
Collapse
MESH Headings
- Adaptation, Physiological/drug effects
- Animals
- Anti-Anxiety Agents/pharmacology
- Brain/cytology
- Brain/drug effects
- Buprenorphine/pharmacology
- Clorazepate Dipotassium/pharmacology
- Drug Interactions
- Male
- Narcotic Antagonists/pharmacology
- Rats
- Rats, Wistar
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, kappa/drug effects
- Receptors, Opioid, kappa/metabolism
- Tissue Distribution
Collapse
Affiliation(s)
- Thomas Quentin
- UMR CEA E2-FRE CNRS 2698 Research Group, Center Cyceron, 15 Boulevard Henry Becquerel, 14070 Caen cedex, France.
| | | | | | | | | | | |
Collapse
|
39
|
Roberts DM, Meyer-Witting M. High-dose buprenorphine: perioperative precautions and management strategies. Anaesth Intensive Care 2005; 33:17-25. [PMID: 15957687 DOI: 10.1177/0310057x0503300104] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Buprenorphine has been in clinical use in anaesthesia for several decades. Recently, the high-dose sublingual formulation (Subutex, Reckitt Benckiser, Slough, U.K.) has been increasingly used as maintenance therapy in opioid dependence, as an alternative to methadone and other pharmacological therapies. Buprenorphine has unique pharmacological properties making it well suited for use as a maintenance therapy in opioid dependence. However, these same properties may cause difficulty in the perioperative management of pain. Buprenorphine is a partial opioid agonist, attenuating the effects of supplemental illicit or therapeutic opioid agonists. As a result of its high receptor affinity, supplemental opioids do not readily displace buprenorphine from the opioid receptor in standard doses. High-dose buprenorphine has an extended duration of action that prolongs both of these effects. The perioperative management of patients stabilized on high-dose buprenorphine and undergoing surgery requires consideration of the likely analgesic requirements. Where possible the buprenorphine should be continued. Pain management should focus on maximizing non-opioid analgesia, local anaesthesia and non-pharmacological techniques. Where pain may not be adequately relieved by these methods, the addition of a full opioid agonist such as fentanyl or morphine at appropriate doses should be considered, accompanied by close monitoring in a high dependency unit. In situations where this regimen is unlikely to be effective, preoperative conversion to morphine or methadone may be an option. Where available, liaison with a hospital-based alcohol and drug service should always be considered.
Collapse
Affiliation(s)
- D M Roberts
- Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland
| | | |
Collapse
|
40
|
Megarbane B, Pirnay S, Borron SW, Trout H, Monier C, Risède P, Boschi G, Baud FJ. Flunitrazepam does not alter cerebral distribution of buprenorphine in the rat. Toxicol Lett 2005; 157:211-9. [PMID: 15917146 DOI: 10.1016/j.toxlet.2005.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2004] [Revised: 02/16/2005] [Accepted: 02/17/2005] [Indexed: 10/25/2022]
Abstract
Deaths have been reported among heroin addicts related to combined buprenorphine and flunitrazepam use. The aim of this study was to determine the existence of a drug-drug interaction during the distribution phase of buprenorphine. Arterial blood gases were measured after intravenous administration of buprenorphine alone (30 mg/kg), flunitrazepam alone (40 mg/kg) or both drugs in rats. Buprenorphine kinetics was studied in plasma and in striatum using cerebral microdialysis, both alone and after rat pretreatment with flunitrazepam. In contrast to buprenorphine or flunitrazepam alone, buprenorphine in combination with flunitrazepam induced a significant, rapid and sustained respiratory depression. Arterial PCO2 was increased at 1.5 min (6.7+/-0.2 versus 5.4+/-0.3 and 5.5+/-0.3 kPa, respectively, P=0.04) (mean+/-S.E.M.), and arterial pH decreased (7.37+/-0.02 versus 7.45+/-0.02 and 7.45+/-0.01, respectively, P=0.03). Plasma buprenorphine kinetics was well described by a three-compartment linear model, with a distribution half-life of 7.4+/-2.7 min and an elimination half-life of 463.9+/-152.3 min. However, neither plasma nor striatal buprenorphine kinetics were significantly altered by pre-administration of flunitrazepam. The adverse interaction between flunitrazepam and buprenorphine cannot be explained by a pharmacokinetic drug-drug interaction during the distribution phase of buprenorphine.
Collapse
Affiliation(s)
- Bruno Megarbane
- INSERM U26, Université Paris VII, Hôpital Fernand Widal, Réanimation Médicale et Toxicologique-INSERM U26, Hôpital Lariboisière, 2 Rue Ambroise Paré, 75010 Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
This review covers beta-phenylethylamines and isoquinoline alkaloids derived from them, including further products of oxidation. condensation with formaldehyde and rearrangement, some of which do not contain an isoquinoline system, together with naphthylisoquinoline alkaloids, which have a different biogenetic origin. The occurrence of the alkaloids, with the structures of new bases, together with their reactions, syntheses and biological activities are reported. The literature from July 2002 to June 2003 is reviewed, with 568 references cited.
Collapse
|
42
|
Yamaoka K, Takakura Y. Analysis Methods and Recent Advances in Nonlinear Pharmacokinetics from In Vitro through In Loci to In Vivo. Drug Metab Pharmacokinet 2004; 19:397-406. [PMID: 15681893 DOI: 10.2133/dmpk.19.397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An attempt has been made to review the nonlinearities in the disposition in vitro, in situ, in loci and in vivo mainly from a theoretical point of view. Parallel Michaelis-Menten and linear (first-order) eliminations are often observed in the cellular uptake, metabolism and efflux of drugs. The well-stirred and parallel-tube models are mainly adopted under steady-state conditions in perfusion experiments, whereas distribution, tank-in-series and dispersion models are often used under nonsteady-state conditions with a pulse input. The analysis of the nonlinear local disposition in loci is reviewed from two points of view, namely an indirect method involving physiologically based pharmacokinetics (PBPK) and a direct (two or three samplings) method using live animals. The nonlinear global pharmacokinetics in vivo is reviewed with regard to absorption, elimination (metabolism and excretion) and distribution.
Collapse
Affiliation(s)
- Kiyoshi Yamaoka
- Department of Biopharmaceutics and Drug Metabolism, School of Graduate Pharmaceutical Science, Kyoto University, Japan.
| | | |
Collapse
|
43
|
Zollner C, Shaqura MA, Bopaiah CP, Mousa S, Stein C, Schafer M. Painful inflammation-induced increase in mu-opioid receptor binding and G-protein coupling in primary afferent neurons. Mol Pharmacol 2003; 64:202-10. [PMID: 12869624 DOI: 10.1124/mol.64.2.202] [Citation(s) in RCA: 152] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Opioids mediate their analgesic effects by activating mu-opioid receptors (MOR) not only within the central nervous system but also on peripheral sensory neurons. The peripheral analgesic effects of opioids are best described under inflammatory conditions (e.g., arthritis). The present study investigated the effects of inflammation on MOR binding and G-protein coupling of full versus partial MOR agonists in dorsal root ganglia (DRG) of primary afferent neurons. Our results show that Freund's complete adjuvant (FCA) unilateral hindpaw inflammation induces a significant up-regulation of MOR binding sites (25 to 47 fmol/mg of protein) on DRG membranes without affecting the affinity of either full or partial MOR agonists. In our immunohistochemical studies, the number of MOR-immunoreactive neurons consistently increased. This increase was mostly caused by small-diameter nociceptive DRG neurons. The full agonist DAMGO induced MOR G-protein coupling in DRG of animals without FCA inflammation (EC50 = 56 nM; relative Emax = 100%). FCA inflammation resulted in significant increases in DAMGO-induced MOR G-protein coupling (EC50 = 29 nM; relative Emax = 145%). The partial agonist buprenorphine hydrochloride (BUP) showed no detectable G-protein coupling in DRG of animals without FCA inflammation; however, partial agonist activity of BUP-induced MOR G-protein coupling was detectable in animals with FCA inflammation (EC50 = 1.6 nM; relative Emax = 82%). In behavioral studies, administration of BUP produced significant antinociception only in inflamed but not in noninflamed paws. These findings show that inflammation causes changes in MOR binding and G-protein coupling in primary afferent neurons. They further underscore the important differences in clinical studies testing peripherally active opioids in inflammatory painful conditions.
Collapse
Affiliation(s)
- C Zollner
- Klinik für Anaesthesiologie und operative Intensivmedizin, Freie Universität Berlin, Universitätklinikum Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Borron SW, Monier C, Risède P, Baud FJ. Flunitrazepam variably alters morphine, buprenorphine, and methadone lethality in the rat. Hum Exp Toxicol 2002; 21:599-605. [PMID: 12507255 DOI: 10.1191/0960327102ht303oa] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Opiates and substitution products are frequently abused, alone and in association with benzodiazepines. While this combination may result in severe respiratory depression and death, the quantitative relationship remains uncertain. We performed randomized, blinded intravenous median lethal dose (MLD) studies in Sprague-Dawley rats of morphine, buprenorphine, and methadone, alone and in combination with intraperitoneal flunitrazepam pretreatment. We employed the up-and-down method, performed in quadruplicate, comparing time to death following opioid injection. Results are expressed as median of four series (extremes). The MLDs of morphine, buprenorphine, and methadone alone were 64.0 (33.6:79.5), 234.6 (168.6:284.4), and 22.5 (19.3:24.1) mg/kg, respectively, and 60.6 (35.2:88.2), 38.4 (30.6:54.0), and 13.0 (9.7:13.8) mg/kg, respectively, after pretreatment with 40 mg/kg flunitrazepam. Times to death for morphine, buprenorphine, and methadone alone were 2.5 (0.8:24), 0.02 (0.0:24), and 2.0 (0.0:24) hours, respectively, and 13.5 (0.0:144), 24.0 (0.0:120), and 0.0 (0.0:24) hours, respectively, after pretreatment with flunitrazepam 40 mg/kg, ip. Flunitrazepam significantly altered methadone (P=0.02) and buprenorphine (P=0.02) but not morphine lethality (P=0.77). Flunitrazepam significantly prolonged time to death only for buprenorphine (P<0.01). Flunitrazepam-opioid drug-drug interactions are more complex than is generally believed. Mechanistic studies of flunitrazepam-opioid lethal interactions are needed.
Collapse
Affiliation(s)
- S W Borron
- INSERM U26, Université Paris 7, Hôpital Fernand Widal, 200 rue du Fg St Denis, Paris 75475, France.
| | | | | | | |
Collapse
|