Startseite Does co-administration of paroxetine change oxycodone analgesia: An interaction study in chronic pain patients
Artikel Öffentlich zugänglich

Does co-administration of paroxetine change oxycodone analgesia: An interaction study in chronic pain patients

  • K.K. Lemberg EMAIL logo , T.E. Heiskanen , M. Neuvonen , V.K. Kontinen , P.J. Neuvonen , M.-L. Dahl und E.A. Kalso
Veröffentlicht/Copyright: 1. Januar 2010
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Oxycodone is a strong opioid and it is increasingly used in the management of acute and chronic pain. The pharmacodynamic effects of oxycodone are mainly mediated by the μ-opioid receptor. However, its affinity for the μ-opioid receptor is significantly lower compared with that of morphine and it has been suggested that active metabolites may play a role in oxycodone analgesia. Oxycodone is mainly metabolized by hepatic cytochrome (CYP) enzymes 2D6 and 3A4. Oxycodone is metabolized to oxymorphone, a potent μ-opioid receptor agonist by CYP2D6. However, CYP3A4 is quantitatively a more important metabolic pathway. Chronic pain patients often use multiple medications. Therefore it is important to understand how blocking or inducing these metabolic pathways may affect oxycodone induced analgesia. The aim of this study was to find out whether blocking CYP2D6 would decrease oxycodone induced analgesia in chronic pain patients.

The effects of the antidepressant paroxetine, a potent inhibitor of CYP2D6, on the analgesic effects and pharmacokinetics of oral oxycodone were studied in 20 chronic pain patients using a randomized, double-blind, placebo-controlled cross-over study design. Pain intensity and rescue analgesics were recorded daily, and the pharmacokinetics and pharmacodynamics of oxycodone were studied on the 7th day of concomitant paroxetine (20 mg/day) or placebo administration. The patients were genotyped for CYP2D6, 3A4, 3A5 and ABCB1.

Paroxetine had significant effects on the metabolism of oxycodone but it had no statistically significant effect on oxycodone analgesia or use of morphine for rescue analgesia. Paroxetine increased the dose-adjusted mean AUC0–12h of oxycodone by 19% (−23 to 113%; P = 0.003), and that of noroxycodone by 100% (5–280%; P < 0.0001) but decreased the AUC0–12 h of oxymorphone by 67% (−100 to −22%; P < 0.0001) and that of noroxymorphone by 68% (−100 to −16%; P < 0.0001).

Adverse effects were also recorded in a pain diary for both 7-day periods (placebo/paroxetine). The most common adverse effects were drowsiness and nausea/vomiting. One patient out of four reported dizziness and headache during paroxetine co-administration, whereas no patient reported these during placebo administration (P = 0.0471) indicating that these adverse effects were due to paroxetine.

No statistically significant associations of the CYP2D6 or CYP3A4/5 genotype of the patients and the pharmacokinetics of oxycodone or its metabolites, extent of paroxetine–oxycodone interaction, or analgesic effects were observed probably due to the limited number of patients studied.

The results of this study strongly suggest that CYP2D6 inhibition does not significantly change oxycodone analgesia in chronic pain patients and that the analgesic activity of oxycodone is mainly due to the parent compound and that metabolites, e.g. oxymorphone, play an insignificant role. The clinical implication of these results is that induction of the metabolism of oxycodone may lead to inadequate analgesia while increased drug effects can be expected after addition of potent CYP3A4/5 inhibitors particularly if combined with CYP2D6 inhibitors or when administered to poor metabolizers of CYP2D6.

1 Introduction

Oral oxycodone is increasingly used to manage chronic pain. Patients needing oxycodone often use several concomitant drugs. Thus, safe and effective use of oxycodone requires knowledge about possible drug interactions.

Oxycodone is a derivate of the opium alkaloid thebaine and it shares many similarities in its molecular structure with codeine and morphine. In humans oxycodone has greater oral bioavailability (60–87%) (Leow et al., 1992; Pöyhiä et al., 1992) than morphine (19–30%) (Osborne et al., 1990) and it undergoes extensive metabolism by CYP enzymes, mainly in the liver. The recovery of oxycodone in urine as unmetabolized oxycodone or its direct conjugates is only 8–14% (Pöyhiä et al., 1992).

Oxycodone is a μ-opioid receptor agonist but it has a lower binding affinity for the μ-opioid receptor than morphine (Chen et al., 1991; Monory et al., 1999; Peckham and Traynor, 2006). Yet, oxycodone has good clinical effectiveness after systemic administration (Silvasti et al., 1998; Nieminen et al., 2009; Kalso et al., 1991; Curtis et al., 1999). Interestingly, its analgesic potency is significantly reduced after spinal administration (Backlund et al., 1997; Yanagidate and Dohi, 2004). Therefore, active metabolites of oxycodone have been suggested to be important for oxycodone analgesia (Kalso et al., 1990).

Oxycodone is N-demethylated by CYP3A4/5 to noroxycodone, which is the main metabolic route (Lalovic et al., 2004). The analgesic properties of noroxycodone have not been studied in humans, but in rodents it shows poor analgesic effect (Weinstein and Gaylord, 1979; Lemberg et al., 2006). Oxymorphone is formed via O-demethylation of oxycodone by CYP2D6, but with a significantly lower rate compared with the formation of noroxycodone (Lalovic et al., 2004). Compared with oxycodone, oxymorphone has about 45-fold higher affinity for the μ-opioid receptor and a higher potency to induce intracellular G-protein activation (Peckham and Traynor, 2006; Lemberg et al., 2006; Thompson et al., 2004; Lalovic et al., 2006). Oxymorphone is used as a potent opioid analgesic in humans and in veterinary medicine (Beaver et al., 1977; Dobbins et al., 2002; Aqua et al., 2007).

Codeine must undergo CYP2D6-mediated O-demethylation to morphine to have an analgesic effect. Therefore, CYP2D6 poor metabolizers have no analgesia from codeine (Dayer et al., 1988; Caraco et al., 1996; Poulsen et al., 1996). The significance of CYP2D6-mediated metabolites in oxycodone analgesia has not been studied in pain patients but other psychomotor effects of oxycodone do not seem to depend on CYP2D6-mediated metabolism (Lalovic et al., 2006; Heiskanen et al., 1998).

Paroxetine is a selective serotonin reuptake inhibitor (SSRI) used in the management of depression and other psychiatric disorders. Paroxetine is mainly metabolized by CYP2D6 and it also acts as a potent inhibitor of CYP2D6, interfering with the metabolism of many clinically used drugs (Brøsen et al., 1991; Bloomer et al., 1992; Sindrup et al., 1992; Laugesen et al., 2005).

We used a placebo-controlled, randomized cross-over design to study the effect of paroxetine (20 mg/day for 7 days) on the pharmacodynamics and pharmacokinetics of oral oxycodone in chronic pain patients. In order to control for the CYP2D6 status the patients were genotyped for CYP2D6. In addition, the patients were genotyped for CYP3A4/5 and ABCB1.

2 Materials and methods

2.1 Study design

The study design is illustrated in Fig. 1. Twenty-eight patients (age range 29–76 years) with stable chronic malignant or non-malignant pain were recruited. Both opioid naïve patients and patients who had been on oxycodone or another opioid were included. Twenty patients (8 males and 12 females, median age of 57.5, range 29–76) completed the study. The demographic data of the patients are given in Table 1. Exclusion criteria were concomitant use of drugs that are potent inhibitors of CYP2D6 activity, clinically relevant renal, hepatic, respiratory or cardiac disease, pregnancy, lactation, alcohol or drug misuse and severe depression or other psychiatric disease. Before entering the study, the following laboratory tests were performed: serum creatinine, serum gamma-glutamyl transpeptidase, aspartate aminotransferase, and alanine aminotransferase. These had to be within±15% of the normal limits in order for the patient to be included.

Figure 1 
              The flow of the patients through the study.
Figure 1

The flow of the patients through the study.

During a run-in period, the patients were titrated to an acceptable level of pain relief (the average pain intensity ≤30 on a 100 mm Visual Analogue Scale (VAS)) with controlled-release oxycodone (Oxycontin®, Mundipharma, Finland) tablets taken twice daily with a 12-h interval. For breakthrough pain the patients were instructed to take oral morphine (morphine hydrochloride 4 mg/ml, Helsinki University Central Hospital Pharmacy, Helsinki, Finland) solution in a dose, which was approximately equivalent to 1/6 of their total daily dose of oxycodone. The equianalgesic dose ratio for oral oxycodone and morphine was assumed to be 2:3 (Heiskanen and Kalso, 1997). Once the patient was on a stable dose of oxycodone and needed not more than two doses of rescue analgesia per day for at least 3 days, the patient was randomized to take either placebo or paroxetine 20 mg (Seroxat® 20 mg, GlaxoSmithKline, Mayenne, France) orally once daily in the morning. Pain intensity (average during the period; VAS 100 mm) was recorded in a pain diary at 8 AM (before the morning dose), at 2 PM and at 8 PM before the evening dose of oxycodone. The doses of rescue medication and adverse effects were recorded daily in the diary. The patient was on the first drug (placebo or paroxetine) for 7 days, after which a 1-week wash out period took place before crossing over to the second treatment-phase. Other medications remained unchanged during the treatment-phases.

Table 1

Patient characteristics.

Patient Gender Age (years) Type of pain Daily dose of oxycodone (mg) Non-opioid co-analgesics (daily dose) Other medications (daily dose) As needed-medications (dose)
1 f 56 Neuropathic 40 Paracetamol 1.5 g Alendronate 70 mg/week, amlodipine 5 mg, furosemide 40 mg, warfarin Lidocain-gel
3 m 76 Ca. pancreas 20 Paracetamol 1.5 g
4 m 68 Ca. pancreas 320 Metoclopramide 30mg, osmotic laxative, sodium picosulphate 22.5 mg
5 m 65 Ca. rectum 40 haloperidol 1.5mg, lactulose 20 ml
6 f 49 CRPS 50 Etoricoxib 90 mg, gabapentin 2400 mg, phenoxybenzamine 20 mg, venlafaxine 75 mg Montelucast 10 μg, estradiolvalerate 1 mg, simvastatin 10mg, esomeprazol 40mg, calsiumcarbonate 1 g, colecalciferol 800IU, alendronate 70 mg (week), amiloride 5 mg, hydrochlorothiazide 50mg, candesartan 16mg, fluticasone 300 μg, ebastine 20 mg, formoterole 48 μg, prednisolone 5 mg, theophylline 900 mg, tiotropium bromide 18 μg Acrivastatine 8mg + pseudoephedrine 60 mg, phenylpropanolamine-HCl, budesonide 100 μg, ipratropium bromide + fenoterole hydrobromide 40/100 μg
7 f 36 Neuropathic 30 Etoricoxib 90 mg, gabapentin 300 mg Ondansetron 4 mg
9 m 58 Neuropathic 80 Allopurinol 100 mg, amlodipine 5 mg, bisoprolol 20 mg, digoxin 0.25 mg, enalapril 20 mg, fluticasone 300 μg, glyburide 3.5 mg, hydrochlortiazide 12.5 mg, metformin 1 g, momethasone 200 μg, rosiglitazone 4 mg, salmeterole 150 μg, simvastatin 20mg, tiotropium bromide 18 μg, warfarin Salbutamol 200 μg
10 f 29 Neuropathic 80 Pregabalin 600mg Amiloride 2.5 mg, budesonide 160 μg, formoterole 4.5 μg, hydrochlortiazide 25 mg, tizanidine 4 mg Osmotic laxative
12 m 65 Neuropathic 100 Gabapentin 2000 mg Alendronate 70 mg/week, bisoprolol 2.5 mg, budesonide 400 μg, diazepam 5 mg, ferrosulphate 100 mg, leflunomide 20 mg, prednisolone 7.5 mg, tamsulosine 0.4 mg, verapamil 160 mg Glyceryl nitrate 0.5 mg, omeprazol 20 mg
14 m 59 Neuropathic 120 Gabapentin 3600 mg, mirtazapin 15 mg Acetosalicylic acid 100mg, atorvastatin 20mg, bisoprolol 2.5 mg, budesonide 320 μg, formoterole 4.5 μg, metformin 750 mg, pantoprazole 40 mg, sodium picosulphate 3.75 mg, tamsulosine 0.4 mg Salbutamol 200 μg
15 m 58 Low back pain 40 Amitriptyline 70mg, gabapentin 3600 mg Enalapril 20 mg, folic acid 5 mg/week, lactulose 20 ml, leflunomide 20mg, methotrexate 10 mg/week, metoprolol 95 mg, prednisolone 5 mg, rosuvastatin 10mg, zolpidem 10 mg
16 f 58 Neuropathic 40 Amitriptyline 37.5 mg, chlordiazepoxide 15 mg, ibuprofen 2400 mg, pregabalin 600 mg Bisoprolol 10mg, levothyroxin 0.1 mg, sulphasalazine 3 g, temazepam 20 mg, tizanidine 12 mg Lactulose 20 ml, zyclizine 10mg
19 m 57 Ischemic pain 80 Paracetamol3g, pregabalin 300 mg Bisoprolol 5 mg, diazepam 20mg, kefalexine 1.5g, Rosuvastatin 10 mg, zopiclone 15 mg -
20 m 38 Neuropathic 80 Amitriptyline 120mg, gabapentin 3600 mg Lactulose 20 ml -
21 f 45 Neuropathic 40 Plantago seed laxative 12 g
23 m 63 Ca. pancreas 40 - Hydroxizine 25 mg, pancreatine 900 mg, tamsulosine 0.4 mg mg Lactulose 20 ml, zyclizine 10mg
24 m 51 Neuropathic 60 Paracetamol 2 g, pregabalin 150mg Acetosalicylic acid 100 mg, bisoprolol 5 mg, furosemide 40mg, montelucaste 10mg, potassium chloride 1 g, ramipril 7.5 mg
25 f 49 Neuropathic 20
27 m 57 Low back pain 40 Amitriptyline 50 mg, gabapentin 3600 mg Bisoprolol 5 mg, glyburide 1.75 mg, hydrochlortiazide 12.5 mg, lisinopril 20 mg, metformin 1.5 g Metoclopramide 10 mg, lactulose 20 ml
28 f 61 Ca. rectum 40 Mirtazapin30mg, paracetamol 4g Sodium picosulphate 3.75 mg Ondansetron 5 mg
  1. CRPS (Complex Regional Pain Syndrome), neuropathic (pain due to nerve injury) and Ca. (carcinoma). The doses of warfarin are not given in the table because of the variance depending on the INR-values.

On the 7th day of both treatment-phases, the patient arrived at the Pain Clinic. The patient did not take either oxycodone or paroxetine/placebo in the morning before coming to the Pain Clinic. A normal light breakfast was allowed at home. Timed blood samples (10 ml each) were drawn from the cannulated forearm vein for the measurement of oxycodone, noroxycodone, oxymorphone, noroxymorphone and paroxetine before taking the tablets (oxycodone + paroxetine/placebo) and at 0.5, 1, 2, 4, 6, 8, 10 and 12 h later. Lunch was served at noon (4 h after drug intake). Pain intensity and relief were assessed on a 100 mm visual analogue scale (VAS) and an 8-point verbal rating scale for pain intensity (VRSpi) and a 5-point verbal rating scale for pain relief (VRSpr) every hour. Also, drug effects were assessed using a 100 mm Modified Drug Effect Scale (Pöyhiä et al., 1991). Adverse effects were reported using a questionnaire. Use of rescue medication was recorded throughout the study. The blood samples were collected to tubes containing ethylenediaminetetra-acetic acid (EDTA). Plasma was separated within 30 min, and stored at −20 °C until analysis. In total, the patients visited the Pain Clinic 5 times during the study (inclusion, start of titration, start of first double-blind-phase and end of each double-blind-phase). During the oxycodone dose titration-phase the patients were contacted by the study nurse or the investigator by telephone every 3rd day until a stable opioid dose was reached.

2.2 Determination of plasma oxycodone, three of its metabolites and paroxetine

Plasma concentrations of oxycodone, oxymorphone, noroxycodone, noroxymorphone, and paroxetine were measured by use of PE SCIEX API 3000 liquid chromatography–tandem mass spectrometry system (Sciex Division of MDS Inc., Toronto, Ontario, Canada). The reversed phase gradient chromatography was performed on an XBridge C18 (2.1 mm × 100 mm, 3.5 μm I.D.) analytical column protected by an XBridge C18 (2.1 mm × 10 mm, 3.5 μm I.D.) guard column (Waters Corp., Milford, MASS). The mobile phase consisted of (channel A) 5 mM ammonium formate (pH 9.4, adjusted with 25% ammonium hydroxide solution) and (channel B) methanol, and the mobile phase flow rate was kept at 180 μL/min. d3-Oxycodone, d3 -oxymorphone, d3 -noroxycodone and venlafaxine served as internal standard. d3 -Noroxycodone was also used as internal standard for noroxymorphone. The mass spectrometer was operated in positive TurboIonSpray® mode and the samples were analyzed via multiple reactant monitoring (MRM) employing the transition of the [M+H]+ precursor ion to a product ion for each analyte and internal standard. The selected ion transitions were as follows: m/z 288 to m/z 213 for noroxymorphone, m/z 302 to m/z 227 for noroxycodone and oxymorphone, m/z 305 to m/z 230 For d3-noroxycodone and d3-oxymorphone, m/z 316 to m/z 241 for oxycodone, 319 to m/z 244 for d3 -oxycodone, m/z 330 to m/z 192 for paroxetine, and m/z 278 t/ m/z 58 for venlafaxine. The limit of quantification was 0.1 ng/ml for oxycodone and oxymorphone, 0.25 ng/ml for noroxycodone, noroxymorphone, and 1.0 ng/ml for paroxetine. The interday coefficient of variation (CV) at the concentrations of 0.1, 5.0 and 100 ng/ml (n = 6) was for oxycodone 13, 3.9, 3.3% and for oxymorphone 9.6, 3.6, 8.3%, respectively. The interday CV at the concentrations of 0.25, 5.0 and 100 ng/ml (n = 6) was for noroxycodone 11, 3.5, 4.2%, and for noroxymorphone 8.4, 7.8, 2.8%. The interday CV for paroxetine at 5.0 and 100 ng/ml (n = 6) was 4.2 and 8.0%, respectively.

2.3 Pharmacokinetics

The pharmacokinetics of oxycodone and its metabolites were evaluated, using plasma drug concentrations adjusted by the daily oxycodone dose (in mg), by peak concentration in plasma (Cmax ) and area under the plasma concentration–time curve from 0 to 12 h (AUC0–12 ). The pharmacokinetic calculations were performed with the program Prism 4.0 (GraphPad Software Inc., San Diego, CA, USA).

2.4 Genotyping methods

Blood samples (10 ml) for genotyping were collected into EDTA vacutainer tubes and kept frozen at −20 °C until analysis. Genomic DNA was isolated from whole blood using the QIAamp® DNA Blood Mini Kit (QIAGEN Ltd.). The CYP2D6 alleles *3, *4, *6, *7, *8 as well as *41 were analyzed using TaqMan® Pre-Developed Assay Reagents for allelic discrimination and the ABI PRISM 7000 Sequence Detection System (Applied Biosystems, Foster City, CA, USA). The CYP2D6*5 allele (total deletion of the gene) was detected by long PCR followed by 1% agarose gel electrophoresis (Hersberger et al., 2000). The CYP2D6 gene duplication, which usually confers ultrarapid metabolism, was detected using long PCR (Steijns and Van Der Weide, 1998). When neither the CYP2D6 variants *3, *4, *5, *6, *7, *8, *41 nor the duplication was detected, the allele was classified as functional CYP2D6*1. Subjects carrying one defective allele together with the functional *1 were classified as heterozygous extensive metabolizers and those with two *1 alleles as homozygous extensive metabolizers. Subjects carrying a gene duplication together with CYP2D6*1 were classified as ultrarapid metabolizers. CYP3A4 detrimental alleles were identified by allele-specific PCR followed by digestion with restriction enzymes, CYP3A4*1B (−290A>G; rs 2740574) and CYP3A4*3 (1437T>C, Met445ThR; rs 4986910) (van Schaik et al., 2000; van Schaik et al., 2001) and CYP3A4*4 allele (352A>G, Ile118Val) (Wang et al., 2005). The CYP3A5*2 (27289C>A, T398N; rs 28365083) and CYP3A5*6 (14690G>A, splicing defect; rs 10264272) alleles were also analyzed by allele-specific PCR followed by digestion with restriction enzymes, as previously described by van Schaik et al. (van Schaik et al., 2000; van Schaik et al., 2001; van Schaik et al., 2002). The presence of the CYP3A5*3 (6986A>G, splicing defect; rs 776746) allele was investigated by TaqManTM allelic discrimination in the ABI PRISM 7000 Sequence Detection System (Mirghani et al., 2006). ABCB1 polymorphisms were analyzed with real-time PCR by TaqMan kits purchased from Applied Biosystems (for 1236C>T, rs1128503, Assay ID: C 7586662 10, for 3435C>T, rs1045642, Assay ID: C 7586657 1, and for 2577G>A/T, rs2032582, Forward Primer GTA AGC AGT AGG GAG TAA CAA AAT AAC ACT, Reverse Primer GAC AAG CAC TGA AAG ATA AGA AAG AAC T, 2677G probe VIC-CCT TCC CAG CAC CT, 2677A probe FAM-CTT CCC AGT ACC TTC, 2677T probe FAM-CTT CCC AGA ACC TT), according to the guidelines of the manufacturer.

Figure 2 
              Pain intensities (a) and pain relief (b) rated on a VAS-scale (mean±SEM, n = 20) during co-administration of placebo or paroxetine (20 mg/day) with oral oxycodone. No statistically significant differences between the phases were found.
Figure 2

Pain intensities (a) and pain relief (b) rated on a VAS-scale (mean±SEM, n = 20) during co-administration of placebo or paroxetine (20 mg/day) with oral oxycodone. No statistically significant differences between the phases were found.

2.5 Ethical considerations

The study was approved by the ethics committee of the Department of Surgery of the Helsinki University Central Hospital and the National Agency for Medicines, Finland. All patients gave a written informed consent.

2.6 Statistical analysis

The paired t-test was used for the statistical analysis of the AUCs of study groups and Fisher’s exact test for the statistical analysis for the association between the observed adverse effects and treatments (placebo or paroxetine) (Prism 4.0, GraphPad Software Inc., San Diego, CA, USA). Analysis of variance (ANOVA) for repeated measures (StatView 5.0.1, SAS Institute, Inc., Cary, NC, USA) was used for comparing the VAS-pain values over time (Fig. 3). P < 0.05 was considered to represent a statistically significant difference.

3 Results

Twenty-eight patients were recruited to the study. Twenty patients completed the whole trial. Their demographic data are given in Table 1.

3.1 Analgesia

During a run-in period before randomization, the patients were titrated to stable pain intensity with twice daily oral controlled-release oxycodone. Oxycodone induced good analgesia in all but four patients who were withdrawn from the study due to inadequate analgesia.

During the double-blind-phase, the patients were instructed to take rescue oral morphine mixture to maintain pain relief. The number of additional morphine administrations per day needed for rescue analgesia did not differ significantly between the placebo (0.84 ± 1.0; mean ± SD) and paroxetine (0.63 ± 0.68) phases of the study. Furthermore, paroxetine did not change the visual analogue scale (VAS) values of pain intensity or pain relief compared with placebo, studied on the 7th day of daily co-administration with oxycodone (Fig. 2a and b). The VAS-pain intensity values indicated more pain in the evenings compared with mornings and noon (P < 0.001) (Fig. 3). CYP2D6, 3A4/5 and ABCB1 genotypes had no significant effect on the VAS-pain intensity values (data not shown).

Figure 3 
              The VAS-pain intensity values in the pain diary, recorded at 8 AM(before the morning dose), 2 PM and 8 PM (before the evening dose) during oxycodone twice daily co-administered with either paroxetine (●) or placebo (○) (mean±SEM). The values show a statistically significant increase in pain intensity in the evenings compared with mornings and noon (P < 0.001) in repeated measures ANOVA when time of the day, the drug (paroxetine vs placebo, n.s.) and study days (1–6, n.s.) are included in the analysis as independent factors.
Figure 3

The VAS-pain intensity values in the pain diary, recorded at 8 AM(before the morning dose), 2 PM and 8 PM (before the evening dose) during oxycodone twice daily co-administered with either paroxetine (●) or placebo (○) (mean±SEM). The values show a statistically significant increase in pain intensity in the evenings compared with mornings and noon (P < 0.001) in repeated measures ANOVA when time of the day, the drug (paroxetine vs placebo, n.s.) and study days (1–6, n.s.) are included in the analysis as independent factors.

3.2 Pharmacokinetics

The plasma concentrations of oxycodone and its three metabolites varied greatly from patient to patient according to the individually titrated oxycodone dose. However, the dose-corrected plasma concentrations were quite similar (Fig. 4). The plasma concentrations of oxycodone and noroxycodone were at a similar level (Fig. 4a and b), being clearly higher than those of noroxymorphone (Fig. 4d) and, in particular, those of oxymorphone (Fig. 4c).

Compared with placebo, paroxetine increased the mean peak (dose-adjusted) plasma concentration (Cmax ) of oxycodone by 26% (range −12 to 130%; P = 0.001) and the mean area under plasma (dose-adjusted) concentration–time curve (AUC0–12 h ) of oxycodone by 19% (range −23 to 113%; P = 0.003) (Fig. 4a, Table 2).

Paroxetine increased the mean (dose-adjusted) Cmax of noroxycodone by 102% (2–267%; P < 0.0001) and its mean AUC0–12 h by 100% (5–280%; P < 0.0001), compared with the corresponding values during the placebo-phase (Fig. 4b, Table 2).

Paroxetine decreased the mean (dose-adjusted) Cmax and AUC0–12 h of oxymorphone by 57% (P < 0.0001) and 67% (P < 0.0001), respectively (Fig. 4c, Table 2). Also the mean (dose-adjusted) Cmax and AUC0–12 h of noroxymorphone were greatly reduced by paroxetine, i.e. by 62% (P < 0.0001) and 68% (P < 0.0001), respectively (Fig. 4d, Table 2).

The mean plasma concentration of paroxetine was quite stable during the (7th) study day in the paroxetine-phase (Fig. 4e). All patients had expected paroxetine concentrations during the paroxetine-phase but none had paroxetine in the plasma samples during the placebo-phase, indicating good compliance in the use of paroxetine.

3.3 Patient genotypes

The CYP2D6, CYP3A4, CYP3A5 and ABCB1 genotypes of the 20 patients who completed the study are given in Table 3. Fourteen patients had two functional CYP2D6 alleles (homozygous extensive metabolizers), four had one functional allele (heterozygous extensive metabolizers) and two had three or more functional alleles, being classified as ultrarapid metabolizers. None of the studied patients were poor metabolizers for CYP2D6.

Table 2

Pharmacokinetic parameters.

Placebo-phase Paroxetine-phase Change from placebo-phase (%), mean (range) P value
Oxycodone
Cmax (ng/ml/mg) 0.70 (±0.22) 0.88 (±0.29) 26(–12to130) 0.001
AUC0-12 h (ng/ml/mg h) 6.49 (±2.62) 7.73 (±3.23) 19 (–23 to 113) 0.003
Noroxycodone
Cmax (ng/ml/mg) 0.45 (±0.19) 0.91 (±0.44) 102 (2.0–267) <0.0001
AUC0-12 h (ng/ml/mg h) 4.38 (±1.97) 8.77 (±3.34) 100 (5.2–280) <0.0001
Oxymorphone
Cmax (ng/ml/mg) 0.019 (±0.01) 0.008 (±0.006) –57 (–100 to 4.0) <0.0001
AUC0-12 h (ng/ml/mg h) 0.i5 (±0.08) 0.049 (±0.049) –67 (–100 to–22) <0.0001
Noroxymorphone
Cmax (ng/ml/mg) 0.10 (±0.005) 0.038 (±0.04) –62 (–90 to -7.4) <0.0001
AUC0-12 h (ng/ml/mg h) 0.97 (±0.49) 0.31 (±0.37) –68 (–100 to -16) <0.0001
  1. Cmax: dose-adjusted maximum concentration (ng/ml/mg); AUC0–12 h: dose-adjusted area under plasma drug concentration–time curve (ng/ml/mg h); (±SD).

Table 3

The genotypes of the 20 patients who completed the study.

Patient no.  CYP2D6 CYP3A4 CYP3A5 ABCB1236C>T ABCB3435C>T ABCB2577G>T or A
1 2 *1/*1 *3/*3 T/T T/T T/T
3 1 *1/*1 *3/*3 C/T T/T G/T
4 2 *1/*1 *3/*3 C/T T/T G/T
5 1 *1/*3 *3/*3 t/t C/T T/T
6 3 or more *1/*1 *3/*3 C/T C/T G/T
7 2 *1/*1 *1/*3 C/C C/C G/A
9 2 *1/*1 *3/*3 C/T C/T G/T
10 2 *1/*1 *3/*3 C/C C/T G/T
12 2 *1/*1 *3/*3 C/C C/C G/G
14 2 *1/*1 *3/*3 C/T C/C G/T
15 2 *1/*1 *3/*3 T/T T/T T/T
16 2 *1/*1 *3/*3 T/T T/T T/T
19 2 *1/*1B *3/*3 C/C C/C G/G
20 3 or more *1/*3 *3/*3 T/T T/T T/T
21 1 *1*1 *3/*3 C/C T/T G/G
23 2 *1/*3 *3/*3 C/C C/C G/G
24 2 *3/*3 *3/*3 T/T C/T T/A
25 1 *3/*3 *3/*3 C/T C/T G/G
27 2 *1/*3 *3/*3 T/T T/T T/T
28 2 *1/*1 *3/*3 C/T C/T G/T
  1. CYP2D6 genotype is expressed as number of functional CYP2D6 genes. Patients with two functional CYP2D6 alleles were classified as homozygous extensive metabolizers, with one functional allele as heterozygous extensive metabolizers and with three or more functional alleles as ultrarapid metabolizers.

Figure 4 
              The dose-adjusted plasma concentrations (ng/ml/mg; mean±SEM, n = 20) of oxycodone, noroxycodone, oxymorphone and noroxymorphone after administration of the same dose of oral oxycodone with either placebo or paroxetine (20 mg/day) for 7 days (a–d). The opioid concentrations are divided by the daily oxycodone dose. The plasma concentrations of paroxetine (ng/ml; mean±SEM, n = 20) on day 7 (e). Time zero refers to the time of drug administration.
Figure 4

The dose-adjusted plasma concentrations (ng/ml/mg; mean±SEM, n = 20) of oxycodone, noroxycodone, oxymorphone and noroxymorphone after administration of the same dose of oral oxycodone with either placebo or paroxetine (20 mg/day) for 7 days (a–d). The opioid concentrations are divided by the daily oxycodone dose. The plasma concentrations of paroxetine (ng/ml; mean±SEM, n = 20) on day 7 (e). Time zero refers to the time of drug administration.

No statistically significant associations of the CYP2D6 or CYP3A4/5 genotype of the patients and the pharmacokinetics of oxycodone or its metabolites, extent of paroxetine–oxycodone interaction, or analgesic effects were observed (data not shown). This may also be related to the limited number of patients studied.

3.4 Adverse effects

During the run-in period, nausea and/or vomiting were reported by 11 of the 20 patients (55%) during both phases of the study. Drowsiness occurred in 13 of the 20 patients (65%) during the paroxetine-phase and in 10 patients (50%) during the placebo-phase. One-fourth of the patients reported dizziness and headache during the paroxetine-phase (P = 0.0471), whereas these adverse effects were not reported during the placebo-phase. Several subjective effects were recorded on the 7th day of both treatment-phases in the Pain Clinic with no statistically significant differences between the phases (Fig. 5).

4 Discussion

This study was performed in chronic pain patients providing clinically relevant information about the pharmacology of oxycodone in “real life”. The chronic pain patients requiring opioid medication were a heterogenous group with multiple medications and various diseases (Table 1). Paroxetine is a potent inhibitor of CYP2D6 and it can potentially be co-administered with oxycodone to treat depression. It was chosen instead of quinidine, another potent blocker of CYP2D6 (Otton et al., 1984), which was considered too risky in out patients. The dose of 20 mg/day paroxetine used in this study was considered not to have any significant analgesic effect. The dose of paroxetine in this study was half of what (40 mg/day) was previously reported to have minor analgesic effect in diabetic neuropathy (Sindrup et al., 1990). SSRIs are not considered effective in chronic pain unless they reduce co-morbid depression or anxiety the treatment of which requires higher doses and a longer duration of treatment than in this study. Inhibition of the CYP2D6-mediated metabolism of oxycodone by paroxetine resulted in a clear pharmacokinetic effect with greatly reduced concentrations of oxymorphone and noroxymorphone. This was not reflected in the pharmacodynamics as the oxycodone induced analgesic effect was only slightly affected by paroxetine. The results are in agreement with the pharmacokinetic studies performed in healthy volunteers (Lalovic et al., 2006; Heiskanen et al., 1998). Thus, the results suggest that oxycodone can be administered with paroxetine or other drugs inhibiting CYP2D6 without interfering with oxycodone analgesia.

All 20 patients completing the study were extensive (18) or ultrarapid (2) metabolizers via CYP2D6, and therefore a suitable population to study CYP2D6-mediated metabolism by blocking its function with paroxetine (Table 3). Analgesia induced by orally administered oxycodone on study day 7 was not significantly influenced by paroxetine (Fig. 2), which caused a moderate increase in plasma oxycodone concentrations and somewhat greater changes in the plasma concentrations of three of its metabolites (Fig. 4). Similarly, neither the pain intensity levels nor the consumption of additional morphine used for rescue analgesia during the study were significantly changed by paroxetine compared with placebo, further suggesting that similar analgesia was obtained. The slight though not significant decrease in the use of additional morphine in the paroxetine-phase (0.63 administrations/day) compared with the placebo-phase (0.84) may be due to the increase of the plasma concentrations of oxycodone in the paroxetine-phase compared with placebo.

Interestingly, the patients reported more pain towards the evenings compared with the mornings and noon (Fig. 3). This is in agreement with a recent study performed in chronic pain patients with painful diabetic neuropathy and postherpetic neuralgia, showing that pain increased throughout the day, without being affected by gabapentin and/or morphine administration (Odrcich et al., 2006). This difference may be explained by more pain related to activity during the day, but also by diurnal variation in the endogenous pain modulating systems (Petraglia et al., 1983). The clinical implication of this finding is that the evening dose of oxycodone should be higher and/or administered earlier.

Paroxetine caused an about 2-fold increase in the plasma concentrations of noroxycodone, the major metabolite of oxycodone in humans, produced by CYP3A4/5 and further metabolized by CYP2D6 (Table 2, Fig. 4b) (Pöyhiä et al., 1992; Lalovic et al., 2004; Heiskanen et al., 1998; Pöyhiä et al., 1991). A similar increase was previously seen after blocking CYP2D6 with quinidine (Heiskanen et al., 1998). Noroxycodone has a poor antinociceptive potency invivo (Weinstein and Gaylord, 1979; Lemberg et al., 2006), because of its low affinity for the μ-opioid receptor compared with oxycodone (Lalovic et al., 2006). Thus, oxycodone induced analgesia seems not to be dependent on noroxycodone.

Figure 5 
            The graded psychodynamic effects on a 0–10 VAS-scale (mean±SEM) during placebo (●) and paroxetine (○) 20mg/day co-administration. No statistically significant differences were found between the two phases.
Figure 5

The graded psychodynamic effects on a 0–10 VAS-scale (mean±SEM) during placebo (●) and paroxetine (○) 20mg/day co-administration. No statistically significant differences were found between the two phases.

Oxymorphone is the most potent μ-opioid receptor agonist of the studied metabolites of oxycodone (Lalovic et al., 2006). However, the plasma concentrations of oxymorphone were very low (Fig. 4c), as has been observed also in other studies in humans (Lalovic et al., 2006; Heiskanen et al., 1998). Furthermore, paroxetine decreased the AUC0–12 h of oxymorphone, to about one-third of the control value (Fig. 4c) without any decrease in analgesia. This suggests a limited role of oxymorphone in the analgesic effect of oral oxycodone in the treatment of chronic pain. However, a more sensitive human experimental pain model where analgesia was studied after oral oxycodone in poor and extensive metabolizers for CYP2D6 indicated some role for oxymorphone (Zwisler et al., 2009).

The AUC0–12 h of plasma noroxymorphone concentrations was also decreased to about one-third by co-administration of paroxetine with oxycodone. Noroxymorphone is mainly formed by the CYP2D6-dependent O-demethylation of noroxycodone (Lalovic et al., 2004). Noroxymorphone is a relatively potent μ-opioid receptor agonist with a 2–3-fold higher affinity for the μ-opioid receptor compared with oxycodone (Chen et al., 1991; Lalovic et al., 2006). Its potency to activate intracellular G-proteins is 2-fold higher than that of oxycodone in the GTPγ[35S] binding assay (Thompson et al., 2004; Lalovic et al., 2006). The central nervous system concentrations of noroxymorphone have been low after intragastric administration of oxycodone to rats, the brain/plasma concentration ratio for noroxymorphone being as low as 0.008 (Lalovic et al., 2006). This is due to the high hydrophilicity (low logD value) of noroxymorphone and hence poor permeability across the lipid-rich blood–brain-barrier.

No significant differences were found between various CYP2D6 or CYP3A4/5 genotypes in the pharmacokinetics or pharmacodynamics of oxycodone. This is not surprising considering the limited number of subjects in most genotype groups. All subjects were extensive or ultrarapid metabolizers via CYP2D6, i.e. during the control-phase oxymorphone and noroxymorphone were formed in these patients at a higher rate than if the patients had been poor CYP2D6 metabolizers. This is important considering the study design and interpretation of the data, because an unchanged analgesic effect of oxycodone by paroxetine in poor CYP2D6 metabolizers would have been expected to be caused by the absence of CYP2D6-mediated metabolites already during the placebo-phase.

If the metabolites of oxycodone do not significantly contribute to analgesia after oral oxycodone in chronic pain patients, how can we explain the discrepancy between the relatively low μ-opioid receptor affinity and efficacy of oxycodone but effective clinical analgesia compared with morphine? Studies in rats have indicated that oxycodone has a significantly better permeability across the blood–brain-barrier compared with morphine (Lalovic et al., 2006; Boström et al., 2006; Boström et al., 2008). The concentrations of oxycodone are 2–6 times higher in the brain compared with plasma whereas the concentrations of oxymorphone are similar in plasma and the CNS. The penetration of noroxymorphone to the CNS is very low (Lalovic et al., 2006; Boström et al., 2006).

P-glycoprotein is an ATP-dependent efflux transport protein that influences the absorption, distribution and excretion of many clinically relevant drugs (Schinkel, 1999). Many opioids, e.g. morphine and methadone are ligands for P-glycoprotein, which actively decreases their CNS concentrations (Letrent et al., 1999; Thompson et al., 2000). A recent study showed a significant reduction in morphine induced pain relief in cancer patients who carried a genotype associated with a poorly functioning μ-opioid receptor (OPRM1, A118G, homozygous G/G) and a well functioning P-glycoprotein (ABCB1, C3435T, homozygous C/C) (Campa et al., 2008). In vitro and in vivo studies have provided conflicting results regarding the interaction of oxycodone with P-glycoprotein (Boström et al., 2005; Hassan et al., 2007).

In the literature, only a few case reports describing interactions between oxycodone and other drugs have been published. Our study suggests that antidepressants that inhibit CYP2D6 do not significantly affect oxycodone analgesia. However, a pharmacodynamic interaction such as serotonin syndrome can result from co-administration of serotonin reuptake inhibitors and opioids (Rosebraucgh et al., 2001; Karunatilake and Buckley, 2006; Rang and Irving, 2008). Lee et al. reported of a patient who had inadequate pain relief after rifampicin, a potent CYP3A4 enzyme inducer, was added to oxycodone medication (Lee et al., 2006). It was suggested that the reduced analgesic efficacy was related to lower oxycodone concentrations due to increased metabolism of oxycodone by rifampicin. Rifampicin has been shown to significantly decrease the plasma concentrations of oxycodone after both oral and intravenous administration with attenuated pharmacological effects (Nieminen et al., 2009). Also, the CYP3A inhibitor voriconazole significantly increased the plasma concentrations of oxycodone with a modest change of some pharmacodynamic effects not including analgesia (Hagelberg et al., 2009).

Our study supports the view that oxycodone induced analgesia and adverse effects are mainly due to the parent drug rather than its metabolites. Thus, induction of the metabolism of oxycodone may lead to inadequate analgesia while increased drug effects can be expected after addition of potent CYP3A4/5 inhibitors as suggested by recent studies by other groups. This could be the case particularly if CYP3A4/5 inhibitors are combined with CYP2D6 inhibitors, leading to increased plasma oxycodone concentrations.


DOI of refers to article: 10.1016/j.sjpain.2009.09.002.



Institute of Biomedicine/Pharmacology, P.O. Box 63, FIN-00014 University of Helsiki, Finland. Tel.: +358 50 5950009; fax: +358 9 19125364.

Acknowledgement

This study received financial support from the Helsinki University Central Hospital Research Funds (T102010066), the Swedish Research Council (521-2005-4771) and an unrestricted grant from Mundipharma, Finland. Eija Kalso has received honoraria for lectures from Egalet, Janssen-Cilag, Mundipharma, Nycomed, Orion Pharma, Pfizer, Prostrakan, and Wyeth. Tarja Heiskanen has received honoraria for lectures from Janssen-Cilag, Mundipharma, and Wyeth. Vesa K. Kontinen has received honoraria for lectures from Janssen-Cilag, MSD, and Pfizer. We wish to thank all patients who participated in the study and Soile Haakana, RN and Leena Murto, RN, Gunilla Frenne, engineer, and Maria Gabriella Scordo, MD, PhD, for excellent assistance in the study.

References

Aqua K, Gimbel JS, Singla N, Ma T, Ahdieh H, Kerwin R. Efficacy and tolerability of oxymorphone immediate release for acute postoperative pain after abdominal surgery: a randomized, double-blind, active- and placebo-controlled, parallel-group trial. Clin Ther 2007;29:1000–12.Suche in Google Scholar

Backlund M, Lindgren L, Kajimoto Y, Rosenberg PH. Comparison of epidural morphine and oxycodone for pain after abdominal surgery. J Clin Anaesth 1997;9:30–5.Suche in Google Scholar

Beaver WT, Wallenstein SL, Houde RW, Rogers A. Comparisons of the analgesic effects of oral and intramuscular oxymorphone and of intramuscular oxymorphone and morphine in patients with cancer. J Clin Pharmacol 1977;17:186–98.Suche in Google Scholar

Bloomer JC, Woods FR, Haddock RE, Lennard MS, Tucker GT. The role of cytochrome P4502D6 in the metabolism of paroxetine by human liver microsomes. Br J Clin Pharmacol 1992;33:521–3.Suche in Google Scholar

Boström E, Simonsson US, Hammarlund-Udenaes M. Oxycodone pharmacokinetics and pharmacodynamics in the rat in the presence of the P-glycoprotein inhibitor PSC833. J Pharm Sci 2005;94:1060–6.Suche in Google Scholar

Boström E, Simonsson US, Hammarlund-Udenaes M. In vivo blood–brain barrier transport of oxycodone in the rat-indications for active influx and implications for PK/PD. Drug Metab Dispos 2006;34:1624–31.Suche in Google Scholar

Boström E, Hammarlund-Udenaes M, Simonsson US. Blood–brain barrier transport helps to explain discrepancies in in vivo potency between oxycodone and morphine. Anesthesiology 2008;108:495–505.Suche in Google Scholar

Brøsen K, Gram LF, Kragh-Sørensen P. Extremely slow metabolism of amitriptyline but normal metabolism of imipramine and desipramine in an extensive metabolizer of sparteine, debrisoquine and mephenytoin. Ther Drug Monit 1991;13:177–82.Suche in Google Scholar

Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of ABCB1/MDR1 and OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther 2008;83:559–66.Suche in Google Scholar

Caraco Y, Sheller J, Wood AJ. Pharmacogenetic determination of the effects of codeine and prediction of drug interactions. J Pharmacol Exp Ther 1996;278:1165–74.Suche in Google Scholar

Chen ZR, Irvine RJ, Somogyi AA, Bochner F. Mu receptor binding of some commonly used opioids and their metabolites. Life Sci 1991;48:2165–71.Suche in Google Scholar

Curtis GB, Johnson GH, Clark P, Taylor R, Brown J, O’Callaghan R, et al. Relative potency of controlled-release oxycodone and controlled-release morphine in a postoperative pain model. Eur J Clin Pharmacol 1999;55:425–9.Suche in Google Scholar

Dayer P, Desmeules J, Leemann T, Striberni R. Bioactivation of the narcotic drug codeine in human liver is mediated by the polymorphic monooxygenase catalyzing debrisoquine 4-hydroxylation (cytochrome P-450 dbl/bufl). Biochem Biophys Res Commun 1988;152:411–6.Suche in Google Scholar

Dobbins S, Brown NO, Shofer FS. Comparison of the effects of buprenorphine, oxymorphone hydrochloride, and ketoprofen for postoperative analgesia after onychectomy or onychectomy and sterilization in cats. J Am Anim Hosp Assoc 2002;38:507–14.Suche in Google Scholar

Hagelberg NM, Nieminen TH, Saari TI, Neuvonen M, Neuvonen PJ, Laine K, et al. Voriconazole drastically increases exposure to oral oxycodone. Eur J Clin Pharmacol 2009;65:263–71.Suche in Google Scholar

Hassan HE, Myers AL, Lee IJ, Coop A, Eddington ND. Oxycodone induces overexpression of P-glycoprotein (ABCB1) and affects paclitaxel’s tissue distribution in Sprague Dawley rats. J Pharm Sci 2007;96:2494–506.Suche in Google Scholar

Heiskanen T, Kalso E. Controlled-release oxycodone and morphine in cancer related pain. Pain 1997;73:37–45.Suche in Google Scholar

Heiskanen T, Olkkola KT, Kalso E. Effects of blocking CYP2D6 on the pharmacokinetics and pharmacodynamics of oxycodone. Clin Pharmacol Ther 1998;64:603–11.Suche in Google Scholar

Hersberger M, Marti-Jaun J, Rentsch K, Hanseler E. Rapid detection of the CYP2D6*3, CYP2D6*4, and CYP2D6*6 alleles by tetra-primer PCR and of the CYP2D6*5 allele by multiplex long PCR. Clin Chem 2000;46:1072–7.Suche in Google Scholar

Kalso E, Vainio A, Mattila MJ, Rosenberg PH, Seppälä T. Morphine and oxycodone in the management of cancer pain: plasma levels determined by chemical and radioreceptor assays. Pharmacol Toxicol 1990;67:322–8.Suche in Google Scholar

Kalso E, Pöyhiä R, Onnela P, Linko K, Tigerstedt I, Tammisto T. Intravenous morphine and oxycodone for pain after abdominal surgery. Acta Anaesthesiol Scand 1991;35:642–6.Suche in Google Scholar

Karunatilake H, Buckley NA. Serotonin syndrome induced by fluvoxamine and oxycodone. Ann Pharmacother 2006;40:155–7.Suche in Google Scholar

Lalovic B, Phillips B, Risler LL, Howald W, Shen DD. Quantitative contribution of CYP2D6 and CYP3A to oxycodone metabolism in human liver and intestinal microsomes. Drug Metab Dispos 2004;32:447–54.Suche in Google Scholar

Lalovic B, Kharasch E, Hoffer C, Risler L, Liu-Chen LY, Shen DD. Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: role of circulating active metabolites. Clin Pharmacol Ther 2006;79:461–79.Suche in Google Scholar

Laugesen S, Enggaard TP, Pedersen RS, Sindrup SH, Brøsen K. Paroxetine, a cytochrome P450 2D6 inhibitor, diminishes the stereoselective Odemethylation and reduces the hypoalgesic effect of tramadol. Clin Pharmacol Ther 2005;77:312–23.Suche in Google Scholar

Lee HK, Lewis LD, Tsongalis GJ, McMullin M, Schur BC, Wong SH, et al. Negative urine opioid screening caused by rifampin-mediated induction of oxycodone hepatic metabolism. Clin Chim Acta 2006;367:196–200.Suche in Google Scholar

Lemberg KK, Kontinen VK, Siiskonen AO, Viljakka KM, Yli-Kauhaluoma JT, Korpi ER, et al. Antinociception by spinal and systemic oxycodone: why does the route make a difference? In vitro and in vivo studies in rats. Anesthesiology 2006;105:801–12.Suche in Google Scholar

Leow KP, Smith MT, Williams B, Cramond T. Single-dose and steady-state pharmacokinetics and pharmacodynamics of oxycodone in patients with cancer. Clin Pharmacol Ther 1992;52:487–95.Suche in Google Scholar

Letrent SP, Pollack GM, Brouwer KR, Brouwer KL. Effects of a potent and specific P-glycoprotein inhibitor on the blood–brain barrier distribution and antinociceptive effect of morphine in the rat. Drug Metab Dispos 1999;27:827–34.Suche in Google Scholar

Mirghani RA, Sayi J, Aklillu E, Allqvist A, Jande M, Wennerholm A, et al. CYP3A5 genotype has significant effect on quinine 3-hydroxylation in Tanzanians, who have lower total CYP3A activity than a Swedish population. Pharmacogenet Genomics 2006;16:637–45.Suche in Google Scholar

Monory K, Greiner E, Sartania N, Sallai L, Pouille Y, Schmidhammer H, et al. Opioid binding profiles of new hydrazone, oxime, carbazone and semicarbazone derivatives of 14-alkoxymorphinans. Life Sci 1999;64:2011–20.Suche in Google Scholar

Nieminen TH, Hagelberg NM, Saari TI, Pertovaara A, Neuvonen M, Laine K, et al. Rifampin greatlyreduces the plasma concentrations of intravenous and oral oxycodone. Anesthesiology 2009;110:1371–8.Suche in Google Scholar

Odrcich M, Bailey JM, Cahill CM, Gilron I. Chronobiological characteristics of painful diabetic neuropathy and postherpetic neuralgia: diurnal pain variation and effects of analgesic therapy. Pain 2006;120:207–12.Suche in Google Scholar

Osborne R, Joel S, Trew D, Slevin M. Morphine and metabolite behavior after different routes of morphine administration: demonstration of the importance of the active metabolite morphine-6-glucuronide. Clin Pharmacol Ther 1990;47:12–9.Suche in Google Scholar

Otton SV, Inaba T, Kalow W. Competitive inhibition of sparteine oxidation in human liver by beta-adrenoceptor antagonists and other cardiovascular drugs. Life Sci 1984;34:73–80.Suche in Google Scholar

Peckham EM, Traynor JR. Comparison of the antinociceptive response to morphine and morphine-like compounds in male and female Sprague–Dawley rats. J Pharmacol Exp Ther 2006;316:1195–201.Suche in Google Scholar

Petraglia F, Facchinetti F, Parrini D, Micieli G, De Luca S, Genazzani AR. Simultaneous circadian variations of plasma ACTH, beta-lipotropin, beta-endorphin and cortisol. Horm Res 1983;17:147–52.Suche in Google Scholar

Poulsen L, Brøsen K, Arendt-Nielsen L, Gram LF, Elbaek K, Sindrup SH. Codeine and morphine in extensive and poor metabolizers of sparteine: pharmacokinetics, analgesic effect and side effects. Eur J Clin Pharmacol 1996;51:289–95.Suche in Google Scholar

Pöyhiä R, Olkkola KT, Seppälä T, Kalso E. The pharmacokinetics of oxycodone after intravenous injection in adults. Br J Clin Pharmacol 1991;32:516–8.Suche in Google Scholar

Pöyhiä R, Seppälä T, Olkkola KT, Kalso E. The pharmacokinetics and metabolism of oxycodone after intramuscular and oral administration to healthy subjects. Br J Clin Pharmacol 1992;33:617–21.Suche in Google Scholar

Rang ST, Irving C. Serotonin toxicity caused by an interaction between fentanyl and paroxetine. Can J Anaesth 2008;55:521–5.Suche in Google Scholar

Rosebraucgh CJ, Flockhart DA, Yasuda SU, Woosley RL. Visual hallucination and tremor induced by sertraline and oxycodone in a bone marrow transplant patient. J Clin Pharmacol 2001;41:224–7.Suche in Google Scholar

Schinkel AH. P-glycoprotein, a gatekeeper in the blood–brain barrier. Adv Drug Deliv Rev 1999;36:179–94.Suche in Google Scholar

Silvasti M, Rosenberg P, Seppälä T, Svartling N, Pitkänen M. Comparison of analgesic efficacy of oxycodone and morphine in postoperative intravenous patient-controlled analgesia. Acta Anaesthesiol Scand 1998;142:576–80.Suche in Google Scholar

Sindrup SH, Gram LF, Brøsen K, Eshoj E, Mogensen EF. The selective serotonin reuptake inhibitor paroxetine is effective in the treatment of diabetic neuropathy symptoms. Pain 1990;42:135–44.Suche in Google Scholar

Sindrup SH, Brøsen K, Gram LF, Hallas J, Sjelbo E, Allen A, et al. The relation between paroxetine and sparteine oxidation polymorphism. Clin Pharmacol Ther 1992;51:278–87.Suche in Google Scholar

Steijns LS, Van Der Weide J. Ultrarapid drug metabolism: PCR-based detection of CYP2D6 gene duplication. Clin Chem 1998;44:914–7.Suche in Google Scholar

Thompson SJ, Koszdin K, Bernards CM. Opiate-induced analgesia is increased and prolonged in mice lacking P-glycoprotein. Anesthesiology 2000;92:1392–9.Suche in Google Scholar

Thompson CM, Wojno H, Greiner E, May EL, Rice KC, Selley DE. Activation of G-proteins by morphine and codeine congeners: insights to the relevance of O-and N-demethylated metabolites at mu- and delta-opioid receptors. J Pharmacol Exp Ther 2004;308:547–54.Suche in Google Scholar

van Schaik RH, de Wildt SN, van Iperen NM, Uitterlinden AG, van den Anker JN, Lindemans J. CYP3A4-V polymorphism detection by PCR-restriction fragment length polymorphism analysis and its allelic frequency among 199 Dutch Caucasians. Clin Chem 2000;46:1834–6.Suche in Google Scholar

van Schaik RH, de Wildt SN, Brosens R, van Fessem M, van den Anker JN, Lindemans J. The CYP3A4*3 allele: is it really rare? Clin Chem 2001;47:1104–6.Suche in Google Scholar

van Schaik RH, van der Heiden IP, van den Anker JN, Lindemans J. CYP3A5 variant allele frequencies in Dutch Caucasians. Clin Chem 2002;48:1668–71.Suche in Google Scholar

Wang A, Yu BN, Luo CH, Tan ZR, Zhou G, Wang LS, et al. Ile118Val genetic polymorphism of CYP3A4 and its effects on lipid-lowering efficacy of simvastatin in Chinese hyperlipidemic patients. Eur J Clin Pharmacol 2005;60:843–8.Suche in Google Scholar

Weinstein SH, Gaylord JC. Determination of oxycodone in plasma and identification of a major metabolite. J Pharm Sci 1979;68:527–8.Suche in Google Scholar

Yanagidate F, Dohi S. Epidural oxycodone or morphine following gynaecological surgery. Br J Anaesth 2004;93:362–7.Suche in Google Scholar

Zwisler ST, Enggaard TP, Noehr-Jensen L, Pedersen RS, Mikkelsen S, Nielsen F, et al. The hypoalgesic effect of oxycodone in human experimental pain models in relation to the CYP2D6 oxidation polymorphism. Basic Clin Pharmacol Toxicol 2009;104:335–44.Suche in Google Scholar

Published Online: 2010-01-01
Published in Print: 2010-01-01

© 2009 Scandinavian Association for the Study of Pain

Artikel in diesem Heft

  1. Editorial
  2. Scandinavian Journal of Pain: A networking and publishing tool for pain researchers and pain clinicians in the Nordic countries
  3. Editorial comments
  4. Pain relief with paravertebral blocks or epidural analgesia? Those who do not know the history of paravertebral blocks are condemned to rediscover the complications
  5. Editorial comments
  6. Investigation of drug–drug interactions and pain—From volunteer studies to randomized controlled trials in patients with chronic pain
  7. Editorial comments
  8. Those who do not know their pain-history will repeat previous errors in pain management
  9. Editorial comments
  10. Fear and catastrophizing thoughts aggravate risks of chronic pain after a fracture
  11. Editorial comments
  12. Important knowledge of pain and phantom experiences after breast surgery and leg- or arm-amputation: Value of qualitative pain research
  13. Editorial comments
  14. Dialectical behavioural therapy for complex chronic pain conditions
  15. Editorial comments
  16. Chronic pain conditions after herniorrhaphy decrease with time, but slowly
  17. Editorial comments
  18. Norwegian patients with chronic pain conditions that can be managed with reasonable cost/benefit now have a legally binding right to treatment in Norway
  19. Review
  20. A systematic review of comparative studies indicates that paravertebral block is neither superior nor safer than epidural analgesia for pain after thoracotomy
  21. Original articles
  22. Does co-administration of paroxetine change oxycodone analgesia: An interaction study in chronic pain patients
  23. Original articles
  24. A personal experience learning from two pain pioneers, J.J. Bonica and W. Fordyce: Lessons surviving four decades of pain practice
  25. Original articles
  26. Pain-related fear, catastrophizing and pain in the recovery from a fracture
  27. Original articles
  28. Adult limb and breast amputees’ experience and descriptions of phantom phenomena—A qualitative study
  29. Original articles
  30. Applying dialectical behavior therapy to chronic pain: A case study
  31. Original articles
  32. Natural course of long-term postherniorrhaphy pain in a population-based cohort
  33. Original articles
  34. National guidelines for evaluating pain—Patients’ legal right to prioritised health care at multidisciplinary pain clinics in Norway implemented 2009
Heruntergeladen am 21.9.2025 von https://www.degruyterbrill.com/document/doi/10.1016/j.sjpain.2009.09.003/html
Button zum nach oben scrollen