Buprenorphine is a semisynthetic partial μ-opioid receptor agonist that appears to provide better analgesia for soft tissue pain than for major bone trauma and surgery.1,2,3 Buprenorphine is widely used in small animal practices in Europe, Australia, and South Africa and is commonly used, particularly in cats, in the United States.4,5,6 Only 1 study7 has been conducted on the pharmacokinetics of buprenorphine in dogs, during which suprapharmacologic doses (0.7 to 2.6 mg/kg) were used, thus limiting applicability of these data to clinical use. Pharmacokinetics of buprenorphine at clinically relevant doses (0.005 to 0.02 mg/kg) in dogs8,9 has not been described. The purpose of the study reported here was to determine the pharmacokinetics of buprenorphine following IV administration in dogs and to establish a clinically relevant dosing regimen.
Materials and Methods
Animals—This study protocol was approved by the Cornell University Institutional Animal Care and Use Committee. Six adult dogs (2 male and 4 female) were studied, which included 2 Golden Retrievers, 2 mixed-breed dogs, 1 Labrador Retriever, and 1 Chesapeake Bay Retriever. No clinically important abnormalities were found on physical examination, CBC determination, and serum biochemical analysis. Mean ± SD age and body weight of dogs was 27 ± 10 months and 31 ± 6.8 kg, respectively.
Food was withheld from all dogs for ≥ 8 hours. One jugular furrow was clipped and a topical anesthetic agent applied.10,a After 30 minutes, dogs were manually restrained; the jugular furrow aseptically prepared; and a 19-gauge, 30.5-cm (12-in) jugular catheterb aseptically inserted in a caudad direction until it was estimated, based on prior measurement, to be positioned within the cranial vena cava. The catheter was secured with tape and a soft padded wrap. This catheter was only used for blood sample collection.
Drug administration and data collection—Buprenorphine HClc (0.015 mg/kg, IV bolus) was administered into a cephalic vein over 5 seconds. Clinical signs typical of opioid administration (eg, vomiting, swallowing, retching, ptyalism, sedation, and recumbency) were recorded at each blood sample collection time. Lengths of clinically apparent effects were recorded based on each dog's recumbency, appropriate response to stimuli, and ability to ambulate normally.
Blood sample collection and handling—Blood samples (3 mL) were collected into evacuated tubes at time 0 prior to drug administration and at 2, 5, 10, 15, 20, 30, 40, 60, 90, 120, 180, 240, 360, 540, 720, 1,080, and 1,440 minutes after drug administration. A standard 3-syringe technique was used to withdraw blood (3 mL) from the jugular catheter to ensure the samples were not diluted. The volume of blood withdrawn was replaced with an equal volume of saline (0.9% NaCl) solution at each collection. All samples were processed within 2 hours after collection. The blood was allowed to clot and then centrifuged for 15 minutes at 560 × g. The serum was collected, transferred into polypropylene serum transport tubes,d and frozen at −70°C until overnight shipment on dry ice to the analytical laboratory.e
Serum sample analysis—Serum samples were analyzed by use of a commercial sodium iodide I 125 radioimmunoassay kit for the quantitative measurement of buprenorphine.f The lower limit of detection delineated by the manufacturer is 0.1 ng/mL, and the kit has extensive cross-reactivity with the N-dealkylated metabolite of buprenorphine.11 In humans, these metabolites are undetectable after a single dose (0.3 mg, IV) or following constant rate infusions ranging from 32 to 239 μg/h.12,13 The assay was validated for canine serum by comparing the predicted to known concentration of control samples based on a standard curve. The standard curve was generated by the addition of known amounts of each drug to canine serum. The lower and upper limits of quantification were 0.2 ng/mL and 8.5 ng/mL, respectively. The coefficient of variation for 3 control samples that spanned the detection range was < 15% for the high-concentration control and ≤ 20% for the low-concentration control. All samples (validation and study) were analyzed in duplicate; the coefficient of variability was based on ≥ 5 sets of control samples.
Pharmacokinetic analysis—For each dog, drug concentration versus time data were subjected to noncompartmental analysis with AUC and AUMC calculated to infinity by use of a log-linear trapezoidal model.g A minimum of 3 data points were used to calculate the kel and the terminal t½ according to the following formula:
From these values, other kinetic variables were calculated as follows:
The Cmax was also recorded. The Ebody was calculated by use of the following formula:
Cardiac output was determined for each dog by use of the following formula:
All pharmacokinetic data were tested for normality by use of a normal probability plot and the Shapiro-Wilk statistic (with P ≤ 0.10 used to indicate nonnormality). The harmonic mean and pseudo SD were calculated for t½. Mean ± SD or median (range) was calculated for the remaining variables.
Results
Clinical effects—Dogs did not react adversely to jugular catheter insertion, and all catheters remained patent for the duration of the study. No dog had behaviors of swallowing, retching, vomiting, or ptyalism. All dogs had signs of sedation for a median (range) duration of 3.5 hours (1.5 to 4 hours) with onset 5 minutes after IV injection of the drug. Four of 6 dogs preferred lateral recumbency during this time; the other 2 preferred sternal or standing positions. Five of 6 dogs were assessed to be clinically normal 6 hours after injection; the sixth dog was difficult to evaluate completely because of its nervous nature. All dogs were able to ambulate without ataxia at this time.
Pharmacokinetics—All data, except Tmax and MRT, were normally distributed and were reported as mean ± SD values; Tmax and MRT were reported as median (range) values. The Cmax achieved in the 6 dogs was 14 ± 2.6 ng/mL; t½ achieved was 270 ± 130 minutes (Table 1). The curvilinear (mean ± SD) concentrationtime plot was determined (Figure 1).
Pharmacokinetic variables for each of the 6 dogs and the summarized data.
Dog | Tmax(min) | Cmax (ng/mL) | AUC (ng•min/mL) | t1/2 (min) | Vdss (L/kg) | Cl (mL/min/kg) | MRT (min) | kel (min−1) | Ebody |
---|---|---|---|---|---|---|---|---|---|
1 | 2 | 16 | 2,783 | 279 | 1.26 | 5.4 | 234 | 0.0025 | 0.059 |
2 | 2 | 17 | 4,222 | 292 | 1.78 | 3.6 | 499 | 0.0024 | 0.036 |
3 | 2 | 11 | 1,804 | 175 | 1.66 | 8.3 | 199 | 0.004 | 0.084 |
4 | 2 | 12 | 2,302 | 174 | 1.34 | 6.5 | 206 | 0.004 | 0.072 |
5 | 2 | 13 | 2,945 | 454 | 1.49 | 5.1 | 293 | 0.0015 | 0.054 |
6 | 5 | 17 | 4,439 | 645 | 2.03 | 3.4 | 600 | 0.0011 | 0.037 |
Mean | 2.5 | 14 | 3,082 | 270* | 1.59 | 5.4 | 338 | 0.0026 | 0.057 |
± SD | 1.2 | 2.6 | 1,048 | 130 | 0.285 | 1.9 | 170 | 0.0012 | 0.019 |
Median | 2 | 14.3 | 2,864.2 | 285.4 | 1.57 | 5.2 | 264 | 0.0025 | - |
Range | 2–5 | 11–17 | 1,804–4,439 | 174–645 | 1.26–2.03 | 3.4–8.3 | 199–600 | 0.0011–0.004 | 0.036–0.084 |
Harmonic mean and pseudo SD.
− = Not applicable.
Discussion
Buprenorphine is a highly lipophilic14 derivative of the morphine alkaloid thebaine and is a member of the 6,14-endo-ethanotetrahydro-oripavine class.15,16 The median lethal dose of buprenorphine HCl following IV administration in dogs is 79 mg/kg,17 and in rats the therapeutic index (median lethal dose/median effective dose) is 12,313.15,18
Traditionally, buprenorphine has been thought of as a partial μ-opioid receptor agonist and a κ-receptor antagonist. Results of a more recent study19 indicate that, with regard to its antinociceptive effect, buprenorphine acts as a full receptor agonist at μ-receptors in rats.19 The μ-effects are responsible for supraspinal analgesia, respiratory depression, and miosis. Buprenorphine might have a wider safety profile, compared with other full μ-receptor agonists, especially regarding respiratory depression.16 Properties of κ-antagonism are some degree of spinal analgesia, dysphoria, and psychomimetic effects.16 Buprenorphine also binds to the D-opioid receptor but with lower affinity than to either the μ- or κ-receptors. The functional importance of this is unknown.16 As an avid binder to the μ-opioid receptor, buprenorphine might displace other μ-receptor agonists given concurrently. As such, it could reverse some of the adverse effects of pure receptor agonists while retaining some analgesia. An additional opioid receptor (the ORL 1 receptor) has been identified. Nociceptin, also referred to as orphanin, is this receptor's natural ligand. Nociceptin acts to substantially decrease dopamine concentrations, either directly as an endogenous antagonist of dopamine transport or by inhibiting γ-aminobutyric acid. Within the CNS, actions of nociceptin are either similar or opposite to those of opioids, depending on their location.20,21,22 Buprenorphine reportedly is a partial receptor agonist at the ORL 1 receptors, whereas its metabolite norbuprenorphine is a full receptor agonist at these receptors, although this is controversial.23,24
Pharmacokinetics of buprenorphine have been evaluated in multiple species.7,13,25,26,27,28 In the summation of previously published data,27 broad similarities were found between the disposition of morphine, buprenorphine, and pethidine in dogs and cats. Following IV administration of buprenorphine at 0.01 mg/kg in cats and of 0.7 mg/kg to 4.8 mg/kg in dogs, t½ was 417 minutes and 1,257 minutes, Vdss was 7.1 L/kg and 5.1 L/kg, and Cl was 17.7 mL/kg/min and 10.5 mL/kg/min for cats and dogs, respectively.7,27 Other than the large differences in doses, several other important differences also were found in these studies.7,27 Measurement of buprenorphine concentrations in the study27 in cats were made by use of the same double-antibody radioimmunoassay used in our study presented here, whereas the original study7 in dogs used high-performance liquid chromatography. Use of the radioimmunoassay does not allow for discrimination between buprenorphine and its metabolites, whereas use of high-performance liquid chromatography does. Buprenorphine is metabolized in the liver by N-dealkylation and glucuronidation, resulting in norbuprenorphine and buprenorphine-3-glucuronide, respectively. In dogs, the highest concentrations of buprenorphine-3-glucuronide are detected within 2 minutes of IV administration of a bolus and the decay of buprenorphine and buprenorphine-3-glucuronide parallel each other in plasma.7 The formation of norbuprenorphine appears to be negligible.7 When buprenorphine-3-glucuronide is administered directly into the duodenum, enterohepatic recirculation does occur, but the lack of free buprenorphine in the plasma indicates that any recirculation of the primary metabolite does not affect the half-life of the drug.7 The metabolite profiles following parenteral administration of buprenorphine are similar in humans and dogs,11 indicating that in dogs, as in humans, the impact of cross-reaction with the metabolite is likely to be minimal after a single dose. Despite binding of norbuprenorphine to the radioimmunoassay antibody, results of a study11 in which a radiotracer was used indicate that little norbuprenorphine is found after a single parenteral administration of buprenorphine in dogs.11 In cats, the metabolic products of buprenorphine are unknown but their ability for glucuronidation is limited.28 In humans, serum drug concentrations do not change in proportion to dose, nor do any subjective and physiologic measurements,29 suggesting that the disposition of buprenorphine cannot be predicted by dose. As such, clinical use should be based on a scientific study that describes the relationship.
The Cmax of buprenorphine in dogs in the study presented here was 14 ± 2.6 ng/mL, which is similar to values of 15.5 ng/mL (7.1 to 26.7 ng/mL) and 12.5 ng/mL (2.55 to 19.4 ng/mL) reported27,28 for cats after IV administration of buprenorphine at 0.01 mg/kg and 0.02 mg/kg, respectively. In our study, Tmax was achieved at the first blood sample collection time (2 minutes) following IV administration of buprenorphine in 5 of 6 dogs. It is a limitation of this study that the first blood sample collection time following IV administration of buprenorphine was at 2 minutes because higher and earlier peaks of Cmax and Tmax may have been missed. Because the purpose of this study was to describe the pharmacokinetics of buprenorphine at a clinically used dose, a higher or earlier Cmax or Tmax are unlikely to make a clinical difference. The AUC, representing the total time course of the drug in the body, was 3,082 ng•min/mL in our study, which is higher than values of AUC previously reported for cats (722 to 2,712 ng•min/mL).27,28 Drug administration was IV in each study, thus removing the influence of absorption following oral administration on AUC. As such, differences between dogs and cats are likely the result of differences in Cl, with Cl being less in dogs. The t½ of 270 minutes for dogs in our study is similar to that found for cats (368 and 417 minutes).27,28
In a previous study,7 the disposition of buprenorphine has been reported for dogs at doses that range from 0.7 to 2.6 mg/kg.7 Substantial differences exist among the disposition values between our study and the earlier study.7 However, care must be taken when comparing data of these 2 studies because of differences in study designs. Some of these differences reflect the reported variable (eg, the AUC is not reported for the different doses in the previous study7). In addition, the standard curve of Garrett and Chandran7 did not span the concentrations of drug detected, control samples were not diluted, and evidence of the lower limit of quantitation was not provided. Also, blood samples were collected from the same catheter into which the drug was administered. As such, failure to remove all drug can cause inaccuracies in measurements of plasma drug concentrations (overestimated) and volume of distribution (underestimated).
The volume of drug cleared per unit time is independent of dose for drugs that follow first-order kinetics.30 However, a recent study in rats reported nonlinear kinetics (ie, zero-order elimination) of buprenorphine, likely indicating nonlinear Cl.26 The free fraction of the drug in plasma is determined by the free concentration of the drug. Therefore, in drugs having nonlinear kinetics, the free fraction is not consistent over a range of concentrations.31 The Ebody is directly related to the properties of the drug. The Ebody is the percentage of the drug cleared by the body during a single passage through the clearing organs. Mean ± SD value of Ebody for our study was 0.057 ± 0.019, indicating a low extraction ratio. For drugs with low extraction ratios, the total body clearance is proportional to the free fraction of the drug in plasma, whereas Cl of the free drug is independent of the free fraction of the drug in plasma. Higher doses of buprenorphine therefore may have a higher free fraction of buprenorphine, resulting in a higher total body clearance.26
The Vdss in this study was less than previously reported for dogs7 or cats.27,28 A high Vdss indicates that the drug is not retained in the plasma, as might occur for drugs distributed to fat, or bound to or sequestered in peripheral tissues. Differences may reflect catheter use but also may reflect dose dependency. Protein-binding of highly protein-bound drugs such as buprenorphine (96%) may become saturated at higher doses.26 This increase is consistent with the phenomenon of concentration-dependent binding to plasma proteins; a larger fraction of the drug is shifted to other tissue compartments from the plasma compartment, which could potentially explain the higher volume of distribution with higher doses.26
Pharmacokinetic data are not the major determining factor of dosing regimens for buprenorphine in dogs. Rather, regimens are based on antinociceptive findings by use of a tooth pulp stimulation technique. In a study32 in dogs,32 on subcutaneous and sublingual administration of buprenorphine (0.02 to 0.04 mg/kg), the nociceptive threshold increased by ≥ 30%, compared with the threshold prior to drug administration.32 The duration of effect for all doses or routes of buprenorphine administration was ≥ 3 hours, but the study32 ended while antinociceptive effects were still in effect, and plasma drug concentrations were not reported. However, in cats, the mean ± SD plasma buprenorphine concentration associated with an increase in the thermal threshold for 10°C after IV administration of buprenorphine was 3.18 ± 1.54 ng/mL.28 Antinociceptive plasma concentrations in dogs and cats are similar for fentanyl at 0.95 and 1.07 ng/mL, respectively.33,34 Assuming pharmacodynamic responses are similar between dogs and cats for buprenorphine, based on pharmacokinetics described in our study, IV administration of buprenorphine may provide analgesia for 3 to 4 hours. Added to this time would be effects that are maintained as a result of slow dissociation between ligand-receptor affinity or biophase equilibrium.
The wide range of t½ values in our study may reflect differences in the extent of redistribution from peripheral tissues or the speed of clearance or cytochrome P450 polymorphism. The terminal half-life after IV administration is the time required for the blood or plasma drug concentration to decrease by 50% after pseudoequilibrium has been reached.35 Because kel (the determinant of t½) is derived from the terminal component of the time concentration curve, that is, after pseudoequilibrium is complete, differences in clearance (ie, the ability of the body to eliminate the drug) are a possible reason for the variability in t½ in our study. Alternatively, the cytochrome P450 system, which is responsible for metabolizing buprenorphine, has been determined to have interethnic variability in expression in humans.36 The implications of this are that standard doses of a drug, when metabolized by such a polymorphic enzyme system, could result in prolonged therapeutic effects, lack of effect, or drug toxicosis.36 If this is true for dogs, it could explain the variability in t½. On the other hand, because our study population consisted of mainly retriever-type dogs, we may have limited the effect of cytochrome P450 variability in drug metabolism. Assuming a direct relationship between antinociceptive effects and plasma drug concentrations, the almost 4-fold variation in t½ (174 to 645 minutes) in individual dogs of our study supports the importance of dosing regimens being designed around patient response rather than predefined dosing intervals.
The long duration of action of buprenorphine as an analgesic has been ascribed to its slow dissociation from the μ-opioid receptor,14,37,h resulting in fewer clinical signs of opioid withdrawal and making it an ideal drug for the management of opioid dependence.16 In 2005, Yassen et al19 found that the slow onset and offset of the antinociceptive effect of buprenorphine in humans are not caused by slow receptor association-dissociation kinetics but rather by slow biophase equilibration kinetics. Biophase equilibration determines the disposition characteristics of an opioid at the effect site and influences the biological effect intensity and the duration of the antinociceptive effect.19 Because of its high lipophilicity, buprenorphine is assumed to readily cross the blood-brain barrier, with the delay in onset of the antinociceptive effect most likely caused by distribution of the drug within the brain parenchyma itself.38,39,40 The decline of buprenorphine concentration from the brain of rats is markedly slower than from plasma (t½ brain = 2.3 hours vs t½ plasma = 1.4 hours). The brain-to-plasma concentration ratio after a single dose of buprenorphine administered IV is 3.0 at 15 minutes and 10.5 at 6 hours after administration.41 This has been confirmed in baboons, which also eliminate buprenorphine slower from the brain than from plasma.42 These findings strongly suggest that the elimination of buprenorphine from the brain is the rate-limiting step in termination of the drug's action.43 In cats, a considerable lag time exists between the peak drug concentration and the dynamic effect, likely the result of biophase equilibration. The thermal threshold increased between 30 and 360 minutes when buprenorphine was given at a dose of 0.02 mg/kg transmucosally, with the maximal effect occurring at 90 minutes, whereas Tmax occurred at 1 to 2 minutes after IV administration.28
The study reported here represents the first in vivo study in which pharmacokinetics of buprenorphine were studied and described at a clinically relevant dose in dogs. Further antinociceptive testing is required to determine the therapeutically effective drug concentration and when it is achieved.
ABBREVIATIONS
AUC | Area under the time-concentration curve to infinity |
AUMC | Area under the first moment curve |
Cl | Plasma clearance |
Cmax | Maximal serum drug concentration |
Ebody | Overall body extraction ratio |
kel | Rate constant associated with the terminal elimination phase |
MRT | Mean residence time |
t½ | Elimination half-life |
Tmax | Time of maximal serum drug concentration |
Vdss | Volume of distribution at steady state |
Lidocaine HCl jelly USP, Akorn Inc, Buffalo Grove, Ill.
Intracath, BD Worldwide, Newark, Del.
Bedford Laboratories, Bedford, Ohio.
Globe Scientific Inc, Paramus, NJ.
Clinical Pharmacology Laboratory, Auburn University, Auburn, Ala.
Double antibody, Diagnostics Products Corp, Los Angeles, Calif.
WinNonlin, Pharsight, Mountain View, Calif.
Wuester M, Herz A. Significance of physiochemical properties of opiates for in vitro testing (abstr), in Proceedings. Int Narc Rec Club Meet. In: Kosterlitz HW, ed. Opiates and endogenous opiate peptides. Amsterdam: Elsevier/North-Holland Biomedical Press, 1976;447.
References
- 1.
Mok MS, Lippmann M, Steen SN. Multidose/observational, comparative clinical analgetic evaluation of buprenorphine. J Clin Pharmacol 1981;21:323–329.
- 2.
Tigerstedt I, Tammisto T. Double-blind, multiple-dose comparison of buprenorphine and morphine in postoperative pain. Acta Anaesthesiol Scand 1980;24:462–468.
- 3.
Wang RI, Johnson RP, Robinson N, et al. The study of analgesics following single and repeated doses. J Clin Pharmacol 1981;21:121–125.
- 4.
Lascelles BD, Capner CA, Waterman-Pearson AE. Current British veterinary attitudes to perioperative analgesia for cats and small mammals. Vet Rec 1999;145:601–604.
- 5.
Watson AD, Nicholson A, Church DB, et al. Use of antiinflammatory and analgesic drugs in dogs and cats. Aust Vet J 1996;74:203–210.
- 6.
Joubert KE. The use of analgesic drugs by South African veterinarians. J S Afr Vet Assoc 2001;72:57–60.
- 7.↑
Garrett ER, Chandran VR. Pharmacokinetics of morphine and its surrogates. X: analyses and pharmacokinetics of buprenorphine in dogs. Biopharm Drug Dispos 1990;11:311–350.
- 8.
Muir WW, Hubbell JAE, Skarda RT, et al. Handbook of veterinary anesthesia. 3rd ed. St Louis: Mosby Inc, 2000.
- 9.
Hendrix P, Hansen B. Acute pain management. In: Bonagura J, ed. Kirk's current veterinary therapy: XIII small animal practice. Philadelphia: WB Saunders, 2000;57–61.
- 10.↑
Woodman PJ. Topical lidocaine-prilocaine versus lidocaine for neonatal circumcision: a randomized controlled trial. Obstet Gynecol 1999;93:775–779.
- 11.↑
Bartlett AJ, Lloyd-Jones JG, Rance MJ, et al. The radioimmunoassay of buprenorphine. Eur J Clin Pharmacol 1980;18:339–345.
- 12.
Hand CW, Baldwin D, Moore RA, et al. Radioimmunoassay of buprenorphine with iodine label: analysis of buprenorphine and metabolites in human plasma. Ann Clin Biochem 1986;23:47–53.
- 13.
Hand CW, Sear JW, Uppington J, et al. Buprenorphine disposition in patients with renal impairment: single and continuous dosing, with special reference to metabolites. Br J Anaesth 1990;64:276–282.
- 14.↑
Hambrook JM, Rance MJ. The interaction of buprenorphine with the opiate receptor: lipophilicity as a determining factor in drug-receptor kinetics. In: Kosterlitz HW, ed. Opiates and endogenous opioid peptides. Amsterdam: Elsevier, 1976;295–301.
- 15.
Cowan A, Lewis JW, Macfarlane IR. Agonist and antagonist properties of buprenorphine, a new antinociceptive agent. Br J Pharmacol 1977;60:537–545.
- 16.↑
Johnson RE, Fudala PJ, Payne R. Buprenorphine: considerations for pain management. J Pain Symptom Manage 2005;29:297–326.
- 17.↑
Registry of toxic effects of chemical substances [database online]. San Leandro, Calif: MDL Information Systems Inc, 2002. Available at: www.mdl.com/products/predictive/rtecs/index.jsp. Accessed Jul 1, 2007.
- 18.
Cowan A, Doxey JC, Harry EJ. The animal pharmacology of buprenorphine, an oripavine analgesic agent. Br J Pharmacol 1977;60:547–554.
- 19.↑
Yassen A, Olofsen E, Dahan A, et al. Pharmacokinetic-pharmacodynamic modeling of the antinociceptive effect of buprenorphine and fentanyl in rats: role of receptor equilibration kinetics. J Pharmacol Exp Ther 2005;313:1136–1149.
- 20.
Darland T, Heinricher MM, Grandy DK. Orphanin FQ/ nociceptin: a role in pain and analgesia, but so much more. Trends Neurosci 1998;21:215–221.
- 21.
Hara N, Minami T, Okuda-Ashitaka E, et al. Characterization of nociceptin hyperalgesia and allodynia in conscious mice. Br J Pharmacol 1997;121:401–408.
- 22.
Reinscheid RK, Nothacker HP, Bourson A, et al. Orphanin FQ: a neuropeptide that activates an opioidlike G protein-coupled receptor. Science 1995;270:792–794.
- 23.
Huang P, Kehner GB, Cowan A, et al. Comparison of pharmacological activities of buprenorphine and norbuprenorphine: norbuprenorphine is a potent opioid agonist. J Pharmacol Exp Ther 2001;297:688–695.
- 24.
Bloms-Funke P, Gillen C, Schuettler AJ, et al. Agonistic effects of the opioid buprenorphine on the nociceptin/OFQ receptor. Peptides 2000;21:1141–1146.
- 25.
Yu S, Zhang X, Sun Y, et al. Pharmacokinetics of buprenorphine after intravenous administration in the mouse. J Am Assoc Lab Anim Sci 2006;45:12–16.
- 26.↑
Gopal S, Tzeng TB, Cowan A. Characterization of the pharmacokinetics of buprenorphine and norbuprenorphine in rats after intravenous bolus administration of buprenorphine. Eur J Pharm Sci 2002;15:287–293.
- 27.↑
Taylor PM, Robertson SA, Dixon MJ, et al. Morphine, pethidine and buprenorphine disposition in the cat. J Vet Pharmacol Ther 2001;24:391–398.
- 28.↑
Robertson SA, Lascelles BD, Taylor PM, et al. PK-PD modeling of buprenorphine in cats: intravenous and oral transmucosal administration. J Vet Pharmacol Ther 2005;28:453–460.
- 29.↑
Harris DS, Mendelson JE, Lin ET, et al. Pharmacokinetics and subjective effects of sublingual buprenorphine, alone or in combination with naloxone: lack of dose proportionality. Clin Pharmacokinet 2004;43:329–340.
- 31.↑
Toutain PL, Bousquet-Melou A. Free drug fraction vs free drug concentration: a matter of frequent confusion. J Vet Pharmacol Ther 2002;25:460–463.
- 32.↑
Skingle M, Tyers MB. Further studies on opiate receptors that mediate antinoception: tooth pulp stimulation in the dog. Br J Pharmacol 1980;70:323–327.
- 33.
Robinson TM, Kruse-Elliott KT, Markel MD, et al. A comparison of transdermal fentanyl versus epidural morphine for analgesia in dogs undergoing major orthopedic surgery. J Am Anim Hosp Assoc 1999;35:95–100.
- 34.
Robertson SA, Taylor PM, Sear JW, et al. Relationship between plasma concentrations and analgesia after intravenous fentanyl and disposition after other routes of administration in cats. J Vet Pharmacol Ther 2005;28:87–93.
- 36.↑
Rogers JF, Nafziger AN, Bertino JS Jr. Pharmacogenetics affects dosing, efficacy, and toxicity of cytochrome P450-metabolized drugs. Am J Med 2002;113:746–750.
- 37.
Boas RA, Villiger JW. Clinical actions of fentanyl and buprenorphine. The significance of receptor binding. Br J Anaesth 1985;57:192–196.
- 38.
Ohtani M, Kotaki H, Sawada Y, et al. Comparative analysis of buprenorphine- and norbuprenorphine-induced analgesic effects based on pharmacokinetic-pharmacodynamic modeling. J Pharmacol Exp Ther 1995;272:505–510.
- 39.
Ebling WF, Lee EN, Stanski DR. Understanding pharmacokinetics and pharmacodynamics through computer stimulation: I. The comparative clinical profiles of fentanyl and alfentanil. Anesthesiology 1990;72:650–658.
- 40.
Bouw MR, Xie R, Tunblad K, et al. Blood-brain barrier transport and brain distribution of morphine-6-glucuronide in relation to the antinociceptive effect in rats–pharmacokinetic/ pharmacodynamic modelling. Br J Pharmacol 2001;134:1796–1804.
- 41.↑
Pontani RB, Vadlamani NL, Misra AL. Disposition in the rat of buprenorphine administered parenterally and as a subcutaneous implant. Xenobiotica 1985;15:287–297.
- 42.↑
Shiue CY, Bai LQ, Teng RR, et al. A comparison of the brain uptake of N-(cyclopropyl[11C]methyl)norbuprenorphine ([11C]buprenorphine) and N-(cyclopropyl[11C]methyl)nordi prenorphine ([11C]diprenorphine) in baboon using PET. Int J Rad Appl Instrum B 1991;18:281–288.
- 43.↑
Yassen A, Olofsen E, Romberg R, et al. Mechanism-based pharmacokinetic-pharmacodynamic modeling of the antinociceptive effect of buprenorphine in healthy volunteers. Anesthesiology 2006;104:1232–1242.