Loading

Journal of Pharmacology and Clinical Toxicology

Parent-Metabolite Pharmacokinetic Models for Tramadol – Tests of Assumptions and Predictions

Research Article | Open Access

  • 1. Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
  • 2. Neonatal Intensive Care Unit, Department of Development and Regeneration, University Hospitals Leuven, Belgium
  • 3. Department of Anaesthesiology, University of Auckland, New Zealand
  • 4. Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, USA
  • 5. Department of Pathology, University of São Paulo, Brazil
  • 6. Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Israel
  • 7. Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, USA
  • 8. School of Pharmacy, Texas Tech University Health Sciences Center, USA
  • 9. Department of Veterinary Sciences, Veterinary Teaching Hospital, University of Pisa, Italy
+ Show More - Show Less
Corresponding Authors
Nick Holford, Department of Pharmacology & Clinical Pharmacology, University of Auckland, Private Bag 92019, Auckland, New Zealand; Tel: +64-9-923-6730 Fax: +64-9-373-7090
Abstract

Allometric principles were used to discern cross-species differences in (±)-tramadol disposition and formation of its primary analgesic metabolite, (±)-O-desmethyl-tramadol (M1). Species differences in formation of M1 may help predict the analgesic effectiveness of tramadol. Tramadol was administered intravenously by a zero-order (constant infusion) process or rapid bolus dose and racemic concentrations of tramadol and M1 measured. Data were pooled to define differences between species (human, rat, cat, dog, goat, donkey and horse). A two-compartment linear disposition model with first-order elimination was used to describe tramadol and M1 disposition. Slow metabolizers were detected in 6% of the population and tramadol clearance to M1 was 16.2% that of extensive metabolizers. Tramadol clearance to M1 was slower and tramadol clearance by other pathways was faster in rats, dogs, and horses compared to humans. There are substantial differences between species in the pharmacokinetics of tramadol and its M1 metabolite, which are not explained by differences in body weight. The hypothesis that volumes of distribution are similar across species was shown not to be true. M1 exposure in the goat, donkey and cat was comparable to humans, which indicates it is likely to be an effective analgesic at typically used doses in these species but not in dogs or horses. 

Citation

Holford S, Allegaert K, Anderson BJ, Kukanich B, Sousa AB, et al. (2014) Parent-Metabolite Pharmacokinetic Models for Tramadol – Tests of Assumptions and Predictions. J Pharmacol Clin Toxicol 2(1):1023.

ABBREVIATIONS

Tramadol: (±)-Tramadol; M1: (±)-O-Desmethyl-Tramadol; CLPM: Clearance to M1; CLPO: Tramadol Clearance by Other Routes; QP: Inter-Compartmental Clearance; CLMO: Clearance of M1; VP1: Central Volume; VP2: Peripheral Volume; QM: M1 Inter-Compartmental Clearance; VM1: Central Volume; VM2: Peripheral Volume; T½: Elimination Half-Life; M5: O-NDidesmethyl-Tramadol; M2: N-Desmethyl-Tramadol; Fm:Fraction of Tramadol Converted to M; Fo: Fraction of Tramadol Eliminated by Other Pathways; HPLC: High Performance Liquid Chromatography; GCMS: Gas Chromatography Mass Spectrometry; LC/MS/MS: HPLC-Coupled Tandem Mass Spectrometry; PPV: Population Parameter Variability; RSE: Relative Standard Error; RUV: Residual Unidentified Variability

INTRODUCTION

Pharmacokinetic models describing concentrations, e.g. in plasma, of parent and metabolite after administration of the parent compound must make assumptions because the system is a priori unidentifiable. Tramadol is a centrally acting racemic analgesic structurally related to morphine that mediates analgesia by multiple mechanisms [1]. The moiety (+)-tramadol and its metabolite (+)-O-desmethyl-tramadol (M1) are weak µ-opioid receptor agonists relative to morphine and the antinociceptive effects of tramadol are attributed to a combination of mechanisms. Along with µ-opioid receptor activity, (+)-tramadol and (+)-M1 stimulate neuronal serotonin efflux while reuptake is inhibited by (+)-tramadol [2,3]. Further analgesia is caused by (-)-tramadol competitively inhibiting noradrenaline reuptake in the spinal cord [4].

Ninety percent of 14C label can be recovered in the urine after oral administration of 14C tramadol to humans [5]. Twelve percent of tramadol and 15% of M1 (expressed as fraction of the tramadol dose) are excreted in the urine unchanged in humans [6]. The mean elimination half-life (T½) is 6 hours and the total clearance following intravenous administration has been reported to be 29 L/h in adult humans [7,8]. In all species, the main tramadol metabolites are M1 and M1 glucuronide and sulfate conjugates, O,N-didesmethyl-tramadol (M5) and M5 conjugates, and N-desmethyl-tramadol (M2). In rats and dogs, only 1% of administered tramadol is excreted unchanged in the urine [5].

O-Demethylation of tramadol to M1, the main analgesic metabolite, is catalyzed by CYP2D6. CYP2D6 polymorphisms have been shown to influence M1 production and its subsequent analgesic effect in humans [9]. (+)-M1 alone has been shown to provide substantial antinociception in rats [10].

The pharmacokinetics of tramadol and M1 after intravenous administration of tramadol have been reported in several adult human studies [8,9,11-13] as well as in dogs [14-16], goats [17], horses [18-21], donkeys [22], cats [23] and rats [24]. Of particular importance to the current analysis, one of these studies observed M1 concentrations after direct intravenous administration of M1, allowing estimation of the volume of distribution of M1 [14]. Data have been pooled from these studies in order to construct a pharmacokinetic model for tramadol and M1. This model has been used to define quantitatively the elimination pathway of tramadol and M1 by comparison to adult humans. The use of allometric principles allows comparison of species differences by normalizing size, which ranges over three orders of magnitude. The assumption that volumes of distribution (such as for M1) are similar across species has been tested using direct estimates from studies in dogs.

MATERIALS AND METHODS

Tramadol was administered intravenously in all studies. All non-human species were fasted for 8 to 12 hours prior to administration of tramadol except cats, which had free access to food during the study (Table 1). Human data were acquired from 57 healthy and 56 post-surgery adults. All non-human species were considered to be in the adult stage of their lifespan. Institutional informed consent and ethical approval was obtained for all studies. Specific details can be found in the original publications. All references to tramadol and its M1 metabolite are to the (±) racemic form. 

Population parameter estimations

A two-compartment (central and peripheral) linear disposition model with zero-order input and first-order elimination fitted the tramadol concentrations from all species combined together more closely than a single compartment model. M1 disposition was also better described by a twocompartment model, with first-order input from the tramadol central compartment and first-order elimination (Figure 1). The model parameters were clearance of tramadol (parent) to M1 (CLPM), tramadol clearance by other routes (CLPO), tramadol inter-compartmental clearance (QP), tramadol central volume (VP1), tramadol peripheral volume (VP2), clearance of M1 (CLMO), inter-compartmental clearance of M1 (QM) and M1 central volume (VM1) and peripheral volume (VM2).

When M1 is not administered directly, the fraction of tramadol converted to M1 (Fm), and the fraction of tramadol eliminated by other pathways (Fo) are unknown. Two different models with distinct assumptions were used to try to distinguish CLPO from CLPM and to identify CLMO and VM:

1. Complete conversion in all species (Fm = 1): Assumes all tramadol is converted to M1. Estimates of CLMO / Fm and VM1 / Fm are species specific. CLPO is assumed to be zero. While it is obvious that this assumption cannot be true (because unchanged tramadol is known to be excreted and other metabolites have been identified), it does permit local identifiability of some key parameters and provides a good description of the time course of concentration.

2. Metabolite volume is the same as in the dog (VM1 = VM1dog): VM1 was estimated in 3 dogs after administration of M1. If the estimate of VM1 / Fm is greater than VM1 estimated in dogs then it may be assumed that VM1 is the same as the dog. If VM1 / Fm is less than VM1 for dogs, then VM1 must be less than the value in dogs but otherwise cannot be identified. The VM1 = VM1dog assumption allows Fm to be identified and CLPO can be distinguished from CLPM [25].

Parameter estimates were obtained using a nonlinear mixed effects approach, which can account for population parameter variability (between and within subjects), residual variability (random effects), and parameter differences predicted by covariates (fixed effects). Parameter estimation was performed using NONMEM version VII level 1.1 with the first-order conditional interaction method. Standard errors of the estimates were obtained by non-parametric bootstrapping [26]. Models were compiled with Intel Visual Fortran version 10.1.029 and executed on an Intel Xeon E5335 Processor with Microsoft Windows 2003 Server Service Pack 2. Model building was based on NONMEM’s objective function and by a visual predictive check [27] with prediction correction [28]. Models were nested and an improvement in the objective function was referred to the chi-squared distribution to assess statistical significance, e.g. an objective function change of 3.84 is significant with Type I error of 0.05 with one additional parameter in the model.

Reported tramadol hydrochloride doses were converted to base tramadol, where 1 mg tramadol hydrochloride is equal to 0.8784 mg of tramadol.

M1 concentrations were converted to tramadol milligram equivalents for a simultaneous parent and metabolite fit using a molecular weight of 249.38 mg mmol-1 for M1 and 263.38 mg mmol-1 for tramadol (molar ratio 0.947). M1 measurements from dog study 20 were excluded from analysis because of contamination with other tramadol metabolites. All other assays are believed to have been selective for M1. Stereoselective concentration measurements were converted to racemic concentrations by summation of stereoisomer concentrations.

Covariate analysis

Fractional differences relative to adult humans were estimated for each population parameter Clearance and volume parameters for tramadol and M1 in all species were standardized to a body weight of 70 kg using an allometric model [29] (Equation 1)

 

where Wi is the weight in the ith individual. Allometric scaling with a PWR exponent of ¾ for clearance and 1 for volume of distribution was employed due to its strong theoretical and empirical basis [30]. Fsize is the allometrically scaled fraction of the standard weight, WSTD.

A mixture model was used to distinguish slow from extensive metabolizers of tramadol on the basis of their phenotype. This method estimates the fraction of all subjects (human and non-human) who appear to be in a slow metabolizer subgroup and the value of CLPM relative to CLPM in extensive metabolizers.

Group parameters were based on fixed effects for clearance using species and size. Equation 2 illustrates how a group value of CLPMGRP is calculated from a standard value of CLPMSTD (adult human 70 kg).

 

Individual parameter estimates (e.g.. CLPMi ) were predicted from the group estimate and the variance of ηi , the random between subject differences in the parameter, using an exponential model (Equation 3):

 

Residual unidentified variability was described using a combined proportional and additive residual error model for each observation prediction with random differences, εPROP, εADD. Between-subject differences in residual error were separately identified for tramadol and M1. The variance of the residual unidentified variability, ηRUV,i, was estimated [31]. This is illustrated for a concentration observation prediction, C, in Equation 4.

Table 1: Summary of studies used and analytical methods.

Publication [8,11,12] [13] [9] [14] [14] [15] [17] [20] [21] [23] [24] [22
Study code 3 5 7 20 21 22 30 40 41 50 60 70
Species Human Human Human Dog Dog Dog Goat Horse Horse Cat Rat Donkey
Number subjects 41 16 56 6 3 6 5 6 6 6 4 12
Dose (mg/kg) of 
tramadol HCL except M1 (base) for 
study 21
100 mg 
bolus 
then, 50 
mg over 
30 min
100 mg bolus 3 over 
18 min
4.4 1 (M1) 4 2 2 5 2 20 2.5 over 3 min
Duration of sampling (h) 24-30.5 48 3 6 4 24 15 24 8 8 5 24
Average observations/subject (tramadol) 15 13 3 8 - 9 7 16 9 12 8 9
Average observations/subject 
(M1)
0 13 3 8 8 6 7 14 7 12 8 6
Analytical method GCMS   LC/MS HPLC Fluorescence 
detection 
[36,37]
  HPLC Fluorescence detection [21] HPLC Ultraviolet detection [17] LC/MS/ MS HPLC Fluorescence detection [21] LC/MS/ MS [23] HPLC Fluorescence detection [38] HPLC Fluorescence detection [22]
Enantiomer       Racemate   Racemate Racemate Racemate Racemate Racemate +/- enantiomer Racemate
LLOQ (tramadol and M1)       No details of assay performance   0.005 mg/L 0.025, 0.010 mg/L 0.001 mg/L 0.005 mg/L 0.001 mg/L 0.0025mg/L 0.005 mg/L
CV%         <4% inter- and intraday 7% interand intraday       7% intra day <15% 7% interday
Mean weight kg (range) 71.1 (58- 98)     9.4
 (7.3-12.4)
  20 (18-23) 47.8 (40.7- 54.4) 402.7 (350-492) 513.5 
(479-545)
4.1 (3.8-
4.4)
0.251 
(0.230-
0.295)
343.5 
(300-380)

 

RESULTS AND DISCUSSION

The first assumption that was tested (Fm = 1) assumed complete conversion of tramadol to M1. All estimates of VM1 / Fm were larger than the VM1 estimated in dogs except in cats (74%) and donkeys (72% of dog VM1). The estimate of VM1 / Fm is always an upper bound on the value for VM1 because Fm must be <=1. This means that the VM1 in cats and donkeys must indeed be smaller than the dog, but in other species the finding of a larger VM1 / Fm could be explained by an additional pathway for tramadol elimination (CLPO) other than formation of M1. In cats either the true VM1 is less than that of dogs, or CLPO is zero.

A second assumption (VM1 = VM1dog) was then tested by assuming VM1 in all species was equal to the dog (except the cat and donkey), which allowed estimation of CLPO. The Fm = 1 assumption was kept for the cat and donkey and VM1 / Fm was estimated separately with CLPO fixed to zero. The VM1 = VM1dog objective function (17343.5) was similar to the Fm = 1 model (17350.8), which confirms the inter-changeability of the Fm = 1 with VM1 = VM1dog assumptions. A major improvement in the objective function (17305.7) was obtained by allowing total tramadol clearance in dog Studies 20 and 21 to be different (4.74 times bigger; 14% bootstrap relative standard error (RSE)) compared to dog Study 22. Removing the mixture model to distinguish two distributions of CLPM from the final model worsened the objective function from 17305.7 to 17319.8. This is a significant (p=0.00085) change for the removal of 2 parameters and provides strong support for the existence of a subgroup of slow metabolizers relative to the rest of the population. The parameter estimates for this model including 2 distributions for clearance and different total tramadol clearance for 2 of the dog studies are shown in Tables 2 to 4.

The visual predictive check plots for tramadol (Figure 2) and M1 (Figure 3) show good agreement between the predicted and observed median and 90% intervals. Parameter estimates for the VM1 = VM1dog model are shown in Tables 2, 3 and 4. The mixture model for identification of M1 metabolizer type estimated that 6.0% (49% bootstrap RSE) of the overall population (human and non-human) were slow metabolizers and that these individuals have 16.2% (24% bootstrap RSE) of the CLPM of extensive metabolizers. All slow metabolizers were human except for 1 horse in study 41.

A fundamental assumption of the modelling of inter-species differences was the appropriateness of the theoretical allometric coefficients of ¾ for clearance and 1 for volume parameters. Although a very wide range of weights were included when considering all species, the use of species-specific parameters means that weight differences are only reflected within each species. The within-species range of weights was relatively small and thus testing if the allometric exponents were different from theoretical values could not be performed with any confidence [29].

After using allometry to account for differences in size, there remain large between-species differences in tramadol and M1 pharmacokinetic parameters. These must be attributed to other factors such as genotype, diet and environment, which are not related to size. Although protein binding changes with pH and carnivorous species tend to have a blood pH lower than that of herbivorous species, tramadol is only 20% protein bound in humans [7] and 15% in dogs [32] so plasma protein binding is not expected to explain the large differences observed.

By assuming the volume of distribution of M1 in the dog is the same as that in other species (except the cat and donkey) it was possible to identify and quantify the clearance of tramadol by other pathways. The mixture model estimate of 6.0% slow metabolizers based on the distribution of CLPM agrees with the fraction of slow CYP2D6 genotypes reported in the literature for humans [33]. Our estimate of the relative clearance of tramadol to its M1 metabolite of 16.2% in slow metabolizers is the only estimate we are aware of because of the impracticality of directly determining this fraction in humans.

The total clearance of tramadol and its elimination by conversion to M1 show marked differences between species (Table 5, Figure 4, Figure 5). The dog is outstanding in having much lower clearance to M1 in Study 22 (CLPM). We have shown that the assumption that the volume of distribution of M1 is similar in all species cannot be true for the cat and the donkey. There are also large within-species differences in volumes of distribution of tramadol that raise further doubts about the assumption that the volume of distribution of M1 is the same in all species.

Two major limitations are recognized in this attempt to describe the pharmacokinetics of tramadol and M1. The first is the necessary assumption that the volume of distribution of M1 is the same in dogs and other species (except the cat and donkey). Unless M1 is administered directly, it is not possible to determine the volume of distribution of M1, though an estimate may be obtained under special conditions [34]. Without knowing (or assuming) this volume, it is impossible to determine the fraction of tramadol that is converted to M1 by a first-order process by only measuring M1 concentrations. The second limitation is the use of racemic concentrations of tramadol and M1, which obscures the different pharmacokinetics of the stereoisomers. This remains a challenge for future studies in those species where only the racemate has been studied.

It is difficult to determine if tramadol has pain-relieving activity in non-human species. Human subjects with the CYP2D6 genotype associated with reduced formation of M1, have worse analgesia [9]. Furthermore, the M1 metabolite is 6 times more potent than tramadol in non-human models of analgesia [35]. If M1 is the main determinant of pain relief, then typical dose rates can be used with species-specific values for CLPM, CLPO and CLMO to predict the M1 average concentration. Comparison of the M1 concentration with those known to be effective in humans can be used to see if dosing rates used in non-human species are likely to be effective. Table 6 shows the predicted M1 average steady state concentrations relative to humans. It seems unlikely that effective pain relief would be achieved in dogs or horses with typically used doses.

Table 2: Human parameter estimates and population parameter variability across all species.

Parameter Description Value (RSE) Units PPV
CLPMextensive Clearance of tramadol to M1 10.5 (13%) L/h/70 kg 0.525
CLPMslow CLPM in slow metabolizers 1.70 (27%) L/h/70 kg 0.059
CLPO Clearance of tramadol by other pathways 18.4 (9%) L/h/70 kg 0.762
QP Inter-compartmental clearance of tramadol 105 (29%) L/h/70 kg 0.647
VP1 Central volume of tramadol 90 (16%) L/70 kg 0.549
VP2 Peripheral volume of tramadol 79 (17%) L/70 kg 0.633
CLMO Clearance of M1 84.2 (10%) L/h/70 kg 0.154
QM Inter-compartmental clearance of M1 274 
(112%)
L/h/70 kg 1.65
VM1 Central volume of M1a 78.9 L/70 kg 0.401
VM2 Peripheral volume of M1 131 (24%) L/70 kg 0.412

Model assumed VM1 was the same as in dogs.
a Fixed to value estimated in dogs administered M1 intravenously.
Abbreviations: PPV: Population Parameter Variability ( sqrt ( NONMEM 
OMEGA estimate ) ); RSE: Relative standard error (bootstrap standard 
error / estimate x 100); M1: (±)-O-desmethyl-tramadol; CLPM:
clearance to M1; CLPO: tramadol clearance by other routes; QP: intercompartmental clearance; CLMO: clearance of M1; VP1: central volume; 
VP2: peripheral volume; QM: M1 inter-compartmental clearance; VM1:
central volume; VM2: peripheral volume

Table 3: Correlation of population parameter variability.

  CLPMextensive CLPO VP1 QP VP2
CLPMextensive 1        
CLPO -0.021 1      
VP1 -0.477 0.863 1    
QP 0.754 0.264 -0.04 1  
VP2 0.888 -0.094 -0.485 0.854 1
  CLMO VM1   QM VM2
CLMO 1   QM 1  
VM 0.886 1 VM2 -0.609 1

Model assumed VM1 was the same as in dogs.
Abbreviations: CLPM: clearance to M1; CLPO: tramadol clearance by other routes; QP: inter-compartmental clearance; 
CLMO: clearance of M1; VP1: central volume; VP2: peripheral volume; QM: M1 inter-compartmental clearance; VM1:
central volume; VM2: peripheral volume

Table 4: Residual unidentified variability (RUV) and population parameter variability.

Parameter Description Value Units PPV
CVCP Proportional error tramadol 0.123 - 0.391
SDCP Additive error tramadol 0.901 mcg/L
CVCM Proportional error M1 0.223 - 0.370
SDCM Additive error M1 0.580 mcg/L

Model assumed VM1 was the same as in dogs. CVCP and CVCM are fractional coefficients of variation. Correlation of PPV RUV tramadol with PPV RUV 
M1 = -0.221.
Abbreviations: PPV: Population Parameter Variability; M1: (±)-O-desmethyl-tramadol

Table 5: Comparison of pharmacokinetic parameters across species.

Parameter Rat Cat Dog study 20, 21 Dog study 22 Goat Donkey Horse study 40 Horse study 41
CLPMextensive 0.658 (35%) 3.5 (19%) 0.517a 0.109 (37%) 1.13 (33%) 1.71 (31%) 0.39 (33%) 0.718 (31%)
CLPO 2.79 (27%) 0 7.39a 1.56 (24%) 4.72 (25%) 0 7.89 (12%) 5.99 (13%)
QP 0.193 (66%) 0.62 (19%) 0.306 (55%) 0.156 (224%) 2.94 (70%) 1.73 (61%) 1.33 (34%) 0.605 (35%)
VP1 2.12 (35%) 1.03 (24%) 2.02 (26%) 0.771 (79%) 0.243 (59%) 0.0405 (111%) 0.669 (25%) 0.983 (27%)
VP2 1.37 (35%) 1.12 (12%) 0.696 (153%) 0.146 (83%) 0.579 (28%) 0.344 (26%) 0.937 (15%) 0.397 (20%)
CLMO 0.394 (45%) 0.389 (29%) 1.13b 0.827 (40%) 0.448 (86%)  1.86 (35%) 3.95 (31%) 0.691 (41%)
QM 0.104 (38%) 0.157 (29%) 2.27b 0.287 (86%) 2.23 (189%) 12.1 (31%) 2.04 (67%) 0.216 (31%)
VM1 1 0.742 (27%) 1b 1 1 0.719 (31%) 1 1
VM2 1 0.742 1 1 1 0.719 1 1

Values are fractional differences relative to human. Relative standard error (bootstrap standard error/estimate x 100) is shown in parentheses. VM1 
and VM2 were assumed to be the same in all species except the donkey which had an estimate of VM1 / Fm and VM2 / Fm that was 0.719 × and the cat 
0.742 × the value of VM1 in the dog (Study 21).
a CLPMextensive and CLPO for dog study 20, 21 calculated from dog study 22 times 4.74 (ratio of total parent metabolite clearance in dog study 20, 21 to 
dog study 22). 
b Estimated from dog study 21 only
Abbreviations: CLPM: clearance to M1; CLPO: tramadol clearance by other routes; QP: inter-compartmental clearance; CLMO: clearance of M1; VP1:
central volume; VP2: peripheral volume; QM: M1 inter-compartmental clearance; VM1: central volume; VM2: peripheral volume

Table 6: Prediction of M1 average steady state concentration relative to human at typical dose rates of tramadol hydrochloride.

Species Reference for Analgesic Dose Dose (mg/kg) Dose Interval (h) Weight (kg) Relative to Human M1 (84 mcg/L)
Human [39]* 2 6 70 100%
Rat [40] 5 6 0.5 60%
Cat [41] 4 6 4 692%
Dog [42] 10 6 15 43.4%
Goat None 2 6 50 68%
Donkey None 2 6 350 221%
Horse study 40 [43]** 2 6 450 0.8%
Horse study 41   2 6 500 10%

Tramadol average concentration = Dose Rate / ( CLPM + CLPO ) 
Rate of conversion to M1 = Tramadol average concentration × CLPM 
M1 average concentration = (Rate of conversion to M1) / CLMO
CLPO and CLMO in cats and donkeys are CLPO / Fm and CLMO / Fm.
*=1 mg/kg/6h produced minimum effective M1 analgesic concentrations in patients with post-operative pain around 50% of those predicted from 2 
mg/kg/6h (84 mcg/L) 
**=2mg/kg intravenous single dose to horses did not produce analgesia

CONCLUSION

There are substantial differences between species in the pharmacokinetics of tramadol and its primary metabolite, which are not explained by differences in body weight. The hypothesis that volumes of distribution are similar across species was shown not to be true. M1 exposure in the goat, donkey and cat was comparable to humans, which indicates it is likely to be an effective analgesic at typically used doses in these species but not in dogs or horses.

ACKNOWLEDGEMENTS

We are grateful to Dr Ulrike Stamer, Dr Rasmus Pedersen and Dr Horst Beier for the use of their data from studies in human adults. Karel Allegaert was supported by the Fund for Scientific Research, Flanders (Fundamental Clinical Investigatorship 1800214N).

REFERENCES

1. Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL, et al. Complementary and synergistic antinociceptive interaction between the enantiomers of tramadol. J Pharmacol Exp Ther. 1993; 267: 331- 340.

2. Raffa RB, Friderichs E, Reimann W, Shank RP, Codd EE, Vaught JL. Opioid and nonopioid components independently contribute to the mechanism of action of tramadol, an ‘atypical’ opioid analgesic. J Pharmacol Exp Ther. 1992; 260: 275-285.

3. Bamigbade TA, Davidson C, Langford RM, Stamford JA. Actions of tramadol, its enantiomers and principal metabolite, O-desmethyltramadol, on serotonin (5-HT) efflux and uptake in the rat dorsal raphe nucleus. Br J Anaesth. 1997; 79: 352-356.

4. Halfpenny DM, Callado LF, Hopwood SE, Bamigbade TA, Langford RM, Stamford JA. Effects of tramadol stereoisomers on norepinephrine efflux and uptake in the rat locus coeruleus measured by real time voltammetry. Br J Anaesth. 1999; 83: 909-915.

5. Lintz W, Erlaçin S, Frankus E, Uragg H. [Biotransformation of tramadol in man and animal (author’s transl)]. Arzneimittelforschung. 1981; 31: 1932-1943.

6. Paar WD, Poche S, Gerloff J, Dengler HJ. Polymorphic CYP2D6 mediates O-demethylation of the opioid analgesic tramadol. Eur J Clin Pharmacol. 1997; 53: 235-239.

7. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004; 43: 879-923.

8. Lintz W, Barth H, Becker R, Frankus E, Schmidt-Böthelt E. Pharmacokinetics of tramadol and bioavailability of enteral tramadol formulations. 2nd communication: drops with ethanol. Arzneimittelforschung. 1998; 48: 436-445.

9. Stamer UM, Lehnen K, Höthker F, Bayerer B, Wolf S, Hoeft A, et al. Impact of CYP2D6 genotype on postoperative tramadol analgesia. Pain. 2003; 105: 231-238.

10. Valle M, Garrido MJ, Pavón JM, Calvo R, Trocóniz IF. Pharmacokineticpharmacodynamic modeling of the antinociceptive effects of main active metabolites of tramadol, (+)-O-desmethyltramadol and (-)-O-desmethyltramadol, in rats. J Pharmacol Exp Ther. 2000; 293: 646-653.

11. Lintz W, Becker R, Gerloff J, Terlinden R. Pharmacokinetics of tramadol and bioavailability of enteral tramadol formulations. 4th communication: drops (without ethanol). Arzneimittelforschung. 2000; 50: 99-108.

12. Lintz W, Beier H, Gerloff J. Bioavailability of tramadol after i.m. injection in comparison to i.v. infusion. Int J Clin Pharmacol Ther. 1999; 37: 175-183.

13. Pedersen RS, Damkier P, Brøsen K. Enantioselective pharmacokinetics of tramadol in CYP2D6 extensive and poor metabolizers. Eur J Clin Pharmacol. 2006; 62: 513-521.

14. KuKanich B, Papich MG. Pharmacokinetics of tramadol and the metabolite O-desmethyltramadol in dogs. J Vet Pharmacol Ther. 2004; 27: 239-246.

15. Giorgi M, Del Carlo S, Saccomanni G, ?ebkowska-Wieruszewska B, Kowalski CJ. Pharmacokinetics of tramadol and its major metabolites  following rectal and intravenous administration in dogs. N Z Vet J. 2009; 57: 146-152.

16. Giorgi M, Del Carlo S, Saccomanni G, ?ebkowska-Wieruszewska B, Kowalski CJ. Pharmacokinetic and urine profile of tramadol and its major metabolites following oral immediate release capsules administration in dogs. Vet Res Commun. 2009; 33: 875-885.

17. de Sousa AB, Santos AC, Schramm SG, Porta V, Górniak SL, Florio JC, et al. Pharmacokinetics of tramadol and o-desmethyltramadol in goats after intravenous and oral administration. J Vet Pharmacol Ther. 2008; 31: 45-51.

18. Cox S, Villarino N, Doherty T. Determination of oral tramadol pharmacokinetics in horses. Res Vet Sci. 2010; 89: 236-241.

19. Dhanjal JK, Wilson DV, Robinson E, Tobin TT, Dirikolu L. Intravenous tramadol: effects, nociceptive properties, and pharmacokinetics in horses. Vet Anaesth Analg. 2009; 36: 581-590.

20. Shilo Y, Britzi M, Eytan B, Lifschitz T, Soback S, Steinman A. Pharmacokinetics of tramadol in horses after intravenous, intramuscular and oral administration. J Vet Pharmacol Ther. 2008; 31: 60-65.

21. Giorgi M, Soldani G, Manera C, Ferrarini PL, Sgorbini M, Saccomanni G. Pharmacokinetics of tramadol and its metabolites M1, M2 and M5 in horses following intravenous, immediate release (fasted/fed) and sustained release single dose administration. J Eq Vet Sci. 2007; 27: 481-488.

22. Giorgi M, Del Carlo S, Sgorbini M, Saccomanni G. Pharmacokinetics of tramadol and its metabolites, M1, M2, and M5 in donkeys after intravenous and oral immediate release single-dose administration. J Eq Vet Sci. 2009; 29: 569-574.

23. Pypendop BH, Ilkiw JE. Pharmacokinetics of tramadol, and its metabolite O-desmethyl-tramadol, in cats. J Vet Pharmacol Ther. 2008; 31: 52-59.

24. Parasrampuria R, Vuppugalla R, Elliott K, Mehvar R. Route-dependent stereoselective pharmacokinetics of tramadol and its active O-demethylated metabolite in rats. Chirality. 2007; 19: 190-196.

25. Evans ND, Godfrey KR, Chapman MJ, Chappell MJ, Aarons L, Duffull SB. An identifiability analysis of a parent-metabolite pharmacokinetic model for ivabradine. J Pharmacokinet Pharmacodyn. 2001; 28: 93- 105.

26. Parke J, Holford NH, Charles BG. A procedure for generating bootstrap samples for the validation of nonlinear mixed-effects population models. Comput Methods Programs Biomed. 1999; 59: 19-29.

27. Holford NHG. The visual predictive check – superiority to standard diagnostic (Rorschach) plots [www.page-meeting.org/?abstract=738]. PAGE 2005; 14 (date accessed 10 January 2014).

28. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO. Predictioncorrected visual predictive checks for diagnosing nonlinear mixed-effects models. AAPS J. 2011; 13: 143-151.

29. Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 2008; 48: 303-332.

30. West GB, Brown JH, Enquist BJ. A general model for the origin of allometric scaling laws in biology. Science. 1997; 276: 122-126.

31. Karlsson MO, Jonsson NE, Wiltse CG, Wade JR. Assumption testing in population pharmacokinetic models: Illustrated with an analysis of moxonidine data from congestive heart failure patients. J Pharmacokinet Biopharm. 1998; 26: 207-246.

32. Vettorato E, Zonca A, Isola M, Villa R, Gallo M, Ravasio G, et al. Pharmacokinetics and efficacy of intravenous and extradural tramadol in dogs. Vet J. 2010; 183: 310-315.

33. Caraco Y. Genes and the response to drugs. N Engl J Med. 2004; 351: 2867-2869.

34. Cheung SY, Majid O, Yates JW, Aarons L. Structural identifiability analysis and reparameterisation (parameter reduction) of a cardiovascular feedback model. Eur J Pharm Sci. 2012; 46: 259-271.

35. Purdue Pharma. Ryzolt product information. 2014.

36. Gan SH, Ismail R. Validation of a high-performance liquid chromatography method for tramadol and o-desmethyltramadol in human plasma using solid-phase extraction. J Chromatogr B Biomed Sci Appl. 2001; 759: 325-335.

37. Nobilis M, Kopecký J, Kvetina J, Chládek J, Svoboda Z, Vorísek V, et al. High-performance liquid chromatographic determination of tramadol and its O-desmethylated metabolite in blood plasma. Application to a bioequivalence study in humans. J Chromatogr A. 2002; 949: 11-22.

38. Mehvar R, Elliott K, Parasrampuria R, Eradiri O. Stereospecific highperformance liquid chromatographic analysis of tramadol and its O-demethylated (M1) and N,O-demethylated (M5) metabolites in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2007; 852: 152-159.

39. Lehmann KA, Kratzenberg U, Schroeder-Bark B, Horrichs-Haermeyer G. Postoperative patient-controlled analgesia with tramadol: analgesic efficacy and minimum effective concentrations. Clin J Pain. 1990; 6: 212-220.

40. Beier H, Garrido MJ, Christoph T, Kasel D, Troconiz IF. Semimechanistic pharmacokinetic/pharmacodynamic modelling of the antinociceptive response in the presence of competitive antagonism: the interaction between tramadol and its active metabolite on microopioid agonism and monoamine reuptake inhibition, in the rat. Pharm Res. 2008; 25: 1789-1797.

41. Pypendop BH, Siao KT, Ilkiw JE. Effects of tramadol hydrochloride on the thermal threshold in cats. Am J Vet Res. 2009; 70: 1465-1470.

42. Kukanich B, Papich MG. Pharmacokinetics and antinociceptive effects of oral tramadol hydrochloride administration in Greyhounds. Am J Vet Res. 2011; 72: 256-262.

43. Seo JP, Son WG, Gang S, Lee I. Sedative and analgesic effects of intravenous xylazine and tramadol on horses. J Vet Sci. 2011; 12: 281- 286.

Received : 14 Feb 2014
Accepted : 26 Feb 2014
Published : 04 Mar 2014
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X