Home Determination of oxycodone and its major metabolites noroxycodone and oxymorphone by ultra-high-performance liquid chromatography tandem mass spectrometry in plasma and urine: application to real cases
Article Publicly Available

Determination of oxycodone and its major metabolites noroxycodone and oxymorphone by ultra-high-performance liquid chromatography tandem mass spectrometry in plasma and urine: application to real cases

  • Flaminia Pantano , Stefano Brauneis , Alexandre Forneris , Roberta Pacifici , Enrico Marinelli , Chrystalla Kyriakou , Simona Pichini EMAIL logo and Francesco Paolo Busardò
Published/Copyright: January 12, 2017

Abstract

Background:

Oxycodone is a narcotic drug widely used to alleviate moderate and severe acute and chronic pain. Variability in analgesic efficacy could be explained by inter-subject variations in plasma concentrations of parent drug and its active metabolite, oxymorphone. To evaluate patient compliance and to set up therapeutic drug monitoring (TDM), an ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) assay was developed and validated for the parent drug and its major metabolites noroxycodone and oxymorphone.

Methods:

Extraction of analytes from plasma and urine samples was obtained by simple liquid-liquid extraction. The chromatographic separation was achieved with a reversed phase column using a linear gradient elution with two solvents: acetic acid 1% in water and methanol. The separated analytes were detected with a triple quadrupole mass spectrometer operated in multiple reaction monitoring (MRM) mode via positive electrospray ionization (ESI).

Results:

Separation of analytes was obtained in less than 5 min. Linear calibration curves for all the analytes under investigation in urine and plasma samples showed determination coefficients (r2) equal or higher than 0.990. Mean absolute analytical recoveries were always above 86%. Intra- and inter-assay precision (measured as coefficient of variation, CV%) and accuracy (measured as % error) values were always better than 13%. Limit of detection at 0.06 and 0.15 ng/mL and limit of quantification at 0.2 and 0.5 ng/mL for plasma and urine samples, respectively, were adequate for the purpose of the present study.

Conclusions:

Rapid extraction, identification and quantification of oxycodone and its metabolites both in urine and plasma by UHPLC-MS/MS assay was tested for its feasibility in clinical samples and provided excellent results for rapid and effective drug testing in patients under oxycodone treatment.

Introduction

The challenge in treating patients with acute or chronic pain has always been to achieve the safe, effective and adequate analgesia. Oxycodone, a semi-synthetic opioid, is a μ-opioid receptor agonist with analgesic effects in several pain conditions (cancer pain, non-malignant chronic pain, and post-surgical acute pain) and it is widely used to relieve from moderate to severe pain [1].

Oxycodone is metabolized by the cytochrome P450 (CYP) enzyme system in the liver and only 10% is excreted unchanged in urine [2]. Oxycodone is N-demethylated by CYP3A4 to noroxycodone, weaker analgesic than the parent drug, whereas the O-demethylation of oxycodone to oxymorphone is mediated by CYP2D6 [3], [4]. This latter metabolite is a powerful analgesic, 14 times more potent than oxycodone and has been speculated to confer the analgesic effect [5].

The involvement of different CYPs in the metabolism, elimination, and bio-activation of oxycodone makes the treatment sensitive to clinically meaningful drug-drug interaction leading to altered analgesic or adverse effects profile [6]. Moreover, oxycodone can also be misused by opioids addicted individuals and care should be taken on the part of healthcare providers to confirm and monitor appropriate use [7].

Several liquid and gas chromatographic methods to quantify plasma and/or urine concentration of oxycodone and its metabolites have been described. Most of these procedures coupled chromatographic separation with electrochemical [8], [9], UV spectrophotometric [10], mass spectrometric [11], [12], [13], [14], and tandem mass spectrometric [15], [16], [17], [18], [19], [20], [21], [22] detection. In general, most of these procedures involve post-column study, and the entire procedure appeared to be complex and time-consuming, either for the analysis of oxycodone alone or in combination with other opioids used in palliative care. Only few published methods used tandem mass spectrometry as a detection method for simultaneous determination of oxycodone and its metabolites in human biological samples [18], [19].

In this study, we developed and validated a simple and fast ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) for the simultaneous determination of oxycodone and its major metabolites in human plasma and urine. The method has been applied to blood and urine samples of patients under oxycodone treatment to prove the reliability and robustness of the method and to preliminary investigate the usefulness of the method as tool for therapeutic drug monitoring (TDM) and eventual relationship between plasma concentrations and clinical outcomes.

Materials and methods

Chemicals and reagents

Oxycodone and oxycodone-d3, oxymorphone and oxymorphone-d3, and noroxycodone and noroxycodone-d3 were supplied by Cerilliant Corporation (Round Rock, TX, USA). β-Glucuronidase (type HP-2 from Helix Pomatia), ultrapure water, and all other reagents of analytical grade were purchased from Sigma-Aldrich (Milan, Italy).

Biological samples

Plasma and urine samples were obtained as discharge material by six hospitalized patients under treatment with different dosages and formulations (Depalgos® and Targin®) of oxycodone, whose biological samples were collected for routine controls. Information on sex and age of the patients, oxycodone formulation and dosage, co-medication, and type of pain to be treated was available for all the patients (Table 1). The patients gave signed consent to the use of their discharged biological samples, which were anonymized with correspondent clinical information in a database before analysis.

Table 1:

Information on sex and age, oxycodone formulation and dosage, co-medication, and type of pain to be treated in the six patients of the study.

Patient codeSex/age, yearsMedicationAdministered doseConcomitant medicationPain type
Patient 1F/51Depalgosa20 mg/dayParoxetineChronic headache
Patient 2F/78Targinb10 mg/2 times/dayPregabalinChronic low back pain
Patient 3F/80Targinb10 mg/dayEnalapril

Acetylsalicylic acid

Pregabalin
Chronic low back pain
Patient 4F/68Depalgosa5 mg/dayPregabalin

Alprazolam

Sertraline
Chronic back pain
Patient 5F/94Targinb20 mg/2 times/dayPregabalin

Duloxetine
Chronic low back pain
Patient 6F/63Targinb60 mg/2 times/dayAcetylsalicylic acid

Clonazepam

Alprazolam

Efavirenz, Nevirapine
Chronic sciatica
  1. aDepalgos: immediate-release oxycodone with paracetamol. bTargin: controlled-release oxycodone with naloxone.

Blood was collected in Vacutainers® EDTA plasma tubes and immediately centrifuged after collection, and the obtained plasma was aliquoted into 0.5-mL plastic tubes. Both plasma and urine samples were frozen at –20°C until analysis. No preservatives were added to specimens.

Drug-free plasma and urine samples were obtained from 20 different healthy donors, analyzed during method validation to exclude any source of chromatographic interferences, and mixed to obtain a homogeneous pool of blank samples to be used for calibration standards and quality control (QC) samples.

Standard solutions, calibrators, and controls

Stock solutions of each analyte (1 mg/mL) were combined and diluted with methanol to set working solutions (5.0, 1.0, and 0.1 μg/mL) and stored at −20°C until analysis. The internal standards (ISs) working solution was used at a concentration of 1.0 μg/mL.

Appropriate amount of working solutions were used to spike pre-checked drug-free plasma and urine samples to prepare five different standards for calibration curve. QC samples at three concentrations (low, medium, high) spanning the linear dynamic ranges of the calibration curves for all the matrices of interest were also daily prepared to be included in each analytical batch to check validation parameters (e.g. accuracy, precision, analytical recovery).

Sample preparation

Aliquots of 0.5 mL plasma and urine were added with appropriate amount of ISs working solution (100 ng for urine and 50 ng for plasma) and diluted with 0.5 mL phosphate buffer, pH 8.4. After 30 s of vortexing, analytes were extracted twice with 1.5-mL aliquots of a chloroform/isopropanol (9:1, v/v) mixture.

After centrifugation at 1500 g for 5 min, the organic phase was evaporated to dryness under a stream of nitrogen and re-dissolved in 100 μL mobile phase (1% acetic acid in water).

An aliquot of urine samples, added with 50 μL of 10,000 units/mL β-glucuronidase, was incubated overnight before extraction, to check for eventual presence of glucuronated metabolites.

UHPLC-MS/MS analysis

Oxycodone, oxymorphone, noroxycodone were measured in plasma and urine samples by UHPLC-MS/MS. The chromatographic separation was achieved with a UPLC Acuity HSS C18 column (2.1×150 mm, 1.8 μm) using a linear gradient elution with two solvents: acetic acid 1% in water (solvent A) and methanol (solvent B). Solvent A was maintained at 85% for the first 1.50 min. It was decreased to 40% from 1.50 to 5.0 min, then increased to 85% from 5.0 to 5.1 min and held at 85% from 5.1 to 10.00 min for re-equilibration. The flow rate was kept constant at 0.35 mL/min during the analysis. The separated analytes were detected with a triple quadrupole mass spectrometer operated in multiple reaction monitoring (MRM) mode via positive electrospray ionization (ESI). The applied ESI conditions were the following: capillary voltage, 1.0 kV; desolvation temperature, 600°C; source temperature, 150°C; cone gas flow rate, 50 L/h; desolvation gas flow rate, 1000 L/h; collision gas flow rate, 0.12 mL/min. MRM transitions, cone voltage, and collision energy as well as retention time (RT) were established for each analyte as listed in Table 2.

Table 2:

UHPLC-MS/MS parameters for the MRM acquisition mode (quantification and confirmation transitions for analytes under investigation).

AnalytesRetention time, minMRM transitions
Quantification (m/z)Confirmation (m/z)CV, VCE, eV
Oxycodone2.74316.21>298.32316.21>256.213020
Oxycodone-d32.72319.21>301.323020
Oxymorphone1.29302.21>284.39302.21>242.213520
Oxymorphone-d31.27305.21>287.393520
Noroxycodone2.92302.28>284.26302.28>187.003520
Noroxycodone-d32.90305.28>287.263520

Validation protocol

Validation protocol applied in the present study included linearity, limits of detection (LOD), and quantification (LOQ), precision, accuracy, selectivity, carryover, matrix effect, recovery, and process efficiency, as elsewhere reported [23]. Validation parameters were calculated from five different daily replicates of QC samples along five subsequent working days. Over-the-curve samples (drug-free samples spiked with concentration of analytes five or 10 times higher than the highest calibration point) were tested for calibration curve fitting, precision, and accuracy once they were properly diluted.

Recovery, matrix effects, and process efficiency were calculated using the experimental protocol suggested by Matuszewski et al. [24]. Set 1 were five replicates of QC material prepared in the mobile phase. Sets 2 and 3 were five replicates of blank matrix samples fortified with QC material after and before extraction, respectively. Matrix effects were determined by dividing mean peak areas of set 2 by set 1 multiplied by 100. Recovery was determined by comparing the mean peak areas of analytes under investigation obtained in set 3 to those in set 2 multiplied by 100. Process efficiency expressed as the ratio of the mean peak area of an analyte spiked before extraction (set 3) to the mean peak area of the same analyte standards (set 1) multiplied by 100. The effect of three freeze-thaw cycles (storage at –20°C) on the compounds stability in urine and plasma was evaluated by repeated analysis (n=3) of QC samples. In addition, mid-term stability test was performed for real samples stored at –20°C. Three replicates of two samples were analyzed once a month during a 6-month period. The stability was expressed as a percentage of the initial concentration (first analyzed batch) of the analytes both in QC and real samples.

Results and discussion

UHPLC-MS/MS method validation

Chromatograms obtained after the extraction of spiked plasma sample and spiked urine sample are shown in Figures 1A and 2A. Separation of analytes was obtained in less than 5 min and column re-equilibration in 10 min.

Figure 1: UHPLC-MS/MS chromatogram of an extract of (A) drug-free plasma spiked with 50.0 ng/mL oxycodone, oxymorphone, and noroxycodone; (B) drug-free plasma sample; (C) plasma sample containing 62.0 ng/mL oxycodone, 0.9 ng/mL oxymorphone, and 32.4 ng/mL noroxycodone.
Figure 1:

UHPLC-MS/MS chromatogram of an extract of (A) drug-free plasma spiked with 50.0 ng/mL oxycodone, oxymorphone, and noroxycodone; (B) drug-free plasma sample; (C) plasma sample containing 62.0 ng/mL oxycodone, 0.9 ng/mL oxymorphone, and 32.4 ng/mL noroxycodone.

Figure 2: UHPLC-MS/MS chromatogram of an extract of (A) drug-free urine spiked with 100.0 ng/mL oxycodone, oxymorphone, and noroxycodone; (B) drug-free urine sample; (C) urine sample containing 444.0 ng/mL oxycodone, 20.6 ng/mL oxymorphone, and 1234.8 ng/mL noroxycodone.
Figure 2:

UHPLC-MS/MS chromatogram of an extract of (A) drug-free urine spiked with 100.0 ng/mL oxycodone, oxymorphone, and noroxycodone; (B) drug-free urine sample; (C) urine sample containing 444.0 ng/mL oxycodone, 20.6 ng/mL oxymorphone, and 1234.8 ng/mL noroxycodone.

Linear calibration curves for all the analytes under investigation in urine and plasma samples showed determination coefficients (r2) equal or higher than 0.990 up to 500 ng/mL plasma and urine samples. LOD and LOQ values calculated for each analyte were adequate for the purpose of the present study (Table 3). The intra- and inter-assay precision (measured as coefficient of variation, CV%) and accuracy (measured as % error) values were always lower than 13% and mean absolute analytical recoveries obtained for the three different QC samples were always above 86% (Table 4). The sensitivity and the analytical recovery obtained by the here described method were both significantly better than those obtained in previously published method using HPLC-MS/MS [18].

Table 3:

Linearity results, LOD, and LOQ values for analytes under investigation.

AnalyteCalibration parametersLODb, ng/mLLOQb, ng/mL
EquationaDetermination coefficients (r2)a
Plasma
 Oxycodoney=0.024−0.0220.9990.060.2
 Oxymorphoney=0.024−0.0120.9960.060.2
 Noroxycodoney=0.025−0.0060.9980.060.2
Urine
 Oxycodoney=0.008−0.0270.9990.150.5
 Oxymorphoney=0.009−0.0420.9980.150.5
 Noroxycodoney=0.008−0.0080.9970.150.5
  1. aMean of three replicates of calibration curves. bMean of five replicates. LOD, limits of detection; LOQ, limits of quantification.

Table 4:

Intra-day (n=5) and inter-day (n=15) precision and accuracy and mean analytical recovery for analytes under investigation.

AnalyteIntra-day precision, CV%Inter-day precision, CV%AccuracyMean analytical recovery, %
Low QCMedium QCHigh QCLow QCMedium QCHigh QCLow QCMedium QCHigh QC
Plasma
 Oxycodone9.18.711.990.012.112.52.19.36.090.0
 Oxymorphone12.010.512.591.712.110.79.04.65.191.7
 Noroxycodone7.86.67.397.29.012.35.45.57.997.2
Urine
 Oxycodone13.06.45.586.36.45.58.16.05.386.3
 Oxymorphone7.42.312.487.62.312.49.43.67.187.6
 Noroxycodone8.47.912.196.07.912.16.55.97.796.0

No additional peaks due to endogenous substances that could have interfered with the detection of the compounds of interest were observed in drug-free samples (Figures 1B and 2B). Blank samples injected after the highest point of the calibration curve did not present any traces of carryover.

No significant ion suppression/enhancement (less than 10% analytical signal suppression due to matrix effect) occurred during chromatographic runs.

Regarding the freeze-thaw stability assays for QC samples, no significant degradation was observed after any of the three freeze-thaw cycles; the differences in concentration compared to the initial concentration were lower that 10%. Similar results (differences always lower than 10%) were obtained in case of mid-term stability test, performed re-analyzing replicates of two real plasma and urine samples once a month for 6-month period, assuring the validity of stored samples analysis.

Real samples

The method described here was applied to real plasma and urine samples collected from patients under treatment with oxycodone to evaluate the robustness and reliability of the method and the eventual applicability to compliance study and TDM. Representative chromatograms obtained following the extraction of real samples containing oxycodone, oxymorphone, and noroxycodone are shown in Figures 1C and 2C, and Table 5 shows the concentrations of oxycodone and its major metabolites obtained in the six different patients together with time interval between drug administration and sample collection and related clinical outcomes.

Table 5:

Time interval between drug administration and sample collection, oxycodone, oxymorphone and noroxycodone concentrations in real plasma and urine samples and related clinical outcomes.

Sample codeTime interval between drug intake and sampling, hPlasma, ng/mLUrine, ng/mLHydrolysed urine, ng/mLOutcomes of treatment
Patient 1
 Oxycodone446.7898.11862.5Resolution of symptoms
 Oxymorphone0.527.0207.1
 Noroxycodone57.83327.63605.5
Patient 2
 Oxycodone1211.2616.51292.0Partial resolution of symptoms
 Oxymorphone<LODa37.4525.9
 Noroxycodone17.75218.05396.9
Patient 3
 Oxycodone1218.0117.7282.3Little improvement – impairment of consciousness
 Oxymorphone<LODa18.0343.8
 Noroxycodone7.586.0101.5
Patient 4
 Oxycodone20 min2.121.734.9No substantial benefit
 Oxymorphone<LODa18.962.4
 Noroxycodone13.1214.2211.0
Patient 5
 Oxycodone562.0444.0721.8Partial resolution of symptoms
 Oxymorphone0.920.6447.5
 Noroxycodone32.41234.81040.9
Patient 6
 Oxycodone1535.31510.61968.3Partial resolution of symptoms
 Oxymorphone1.4115.43622.97
 Noroxycodone87.320829.825323.0
  1. a<LOD: under the limit of detection.

When analyte concentration in urine samples was higher than those in the calibration curve, samples were re-processed once diluted 5 or 10× (over-curve samples).

Following oral administration, oxycodone was excreted in the urine primarily as noroxycodone (concentration range, 86.0–20829.8 ng/mL), secondarily as parent drug (concentration range, 21.7–1510.6 ng/mL) and in lower concentration as oxymorphone (concentration range, 18.0–115.4 ng/mL). With respect to substances glucuronidation, in accordance with the study of Fang et al. [18], oxycodone and oxymorphone were primarily excreted in their glucuronide form. Indeed, after urine hydrolysis, oxycodone and oxymorphone concentrations showed about 2× and 10× concentration increase, respectively. The latter did not apply to noroxycodone.

Since oxycodone is widely abused [18], [25] and several fatalities have been attributed to this substance [26], [27], [28], [29], [30], [31], [32], urine analysis could provide a valuable tool to determine the use of this substance.

In five out of the six patients, plasma oxycodone concentrations were found in the range of 11.2–62.0 ng/mL. This concentration range falls within the therapeutic range reported elsewhere (i.e. 5–100 ng/mL) [33]. Only one patient (patient 4) had a concentration of oxycodone below the therapeutic range (i.e. 2.1 ng/mL), suggesting non-compliance. Indeed, when questioned about possible non compliance the patient referred to have taken the drug 20 min before the blood collection in order not to fail the therapy.

The analgesic effects of opioids are related to their plasma concentration. Either mean effective concentration (MEC) or minimum effective analgesic concentration (MEAC) is used to assess concentration-effect relationships.

Oxycodone treatment was effective or partially effective in some of the patients here tested whereas no substantial benefit was observed in others (Table 5). However, with these few data correlation between plasma concentration and clinical outcomes could not be established.

Generally speaking, the relationship between plasma oxycodone concentration and the analgesic response depends on the patients age, state of health, medical condition and the eventual extent of previous opioid treatment [34]. Moreover, variability in analgesic efficacy could be explained by inter-subject variations in plasma levels of parent drug and its active metabolite eventually affected also by co-medications [19], and therefore, dosage must be titrated for optimal effect and avoidance of toxicity after appropriate TDM.

The MEAC of oxycodone to achieve analgesia widely varies among patients, especially among patients who have been previously treated with potent agonist opioids. Thus, patients need to be treated with individualized titration of dosage to reach the desired effect. Moreover, repeated dosing due to an increase in pain and/or development of tolerance, may also increase the MEAC of this substance.

In any case, these were just preliminary data obtained in a very low number of patients with the principal aim of validating a simple and fast UHPLC assay to be applied in clinical TDM of oxycodone treatment.

It is very important for clinical practice to evaluate the compliance to the prescribed dose and to prevent abuse phenomena especially in outpatients cases, taking into consideration that adverse reactions and fatal outcomes can occur. Therefore, it is important to have appropriate diagnostic tools, like the one here described, to manage the therapeutic use of oxycodone as well as to identify potential diversion of therapy and abuse. The main limitation of this study is the small sample size tested and the heterogeneity of the cases (different doses, formulations, co-medications). However, a research on the same topic is ongoing and a larger sample size is to be analyzed to assess how co-medication (CYP2D6 and CYP3A4 inhibitors), dose and formulation can affect the efficacy of the drug on pain relief.

Conclusions

An assay including a simple biological sample treatment and fast UHPLC-MS/MS method was developed for the determination of oxycodone and of its major metabolites in conventional matrices. The method was fully validated and applied to six patients under oxycodone treatment.

The current method could find application in both clinical and forensic toxicology analysis; in emergency rooms and workplace drug testing. Even if it is not an automated analysis and requires a very advanced hyphenated technique, the complete assay is sufficiently easy and simple to be applied in a high-throughput laboratory dealing with drug monitoring, compliance, and pharmacokinetic studies.


Corresponding author: Dr. Simona Pichini, Drug Abuse and Doping Unit, Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, V.le Regina Elena 299, 00161 Rome, Italy, Phone: +39 06 49906545, Fax: +39 06 49902016

Acknowledgments

The authors thank Michele Sciotti, Simonetta di Carlo and Antonella Bacosi for technical assistance.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Kalso E. Oxycodone. J Pain Symptom Manage 2005;29(Suppl): S47–56.10.1016/j.jpainsymman.2005.01.010Search in Google Scholar

2. Poyhia R, Seppala T, Olkkola KT, Kalso E. The pharmacokinetics and metabolism of oxycodone after intramuscular and oral administration to healthy subjects. Br J Clin Pharmacol 1992;33:617–21.10.1111/j.1365-2125.1992.tb04090.xSearch in Google Scholar

3. 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;79:461–79.10.1016/j.clpt.2006.01.009Search in Google Scholar

4. 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;32:447–54.10.1124/dmd.32.4.447Search in Google Scholar

5. Elder NM, Atayee RS, Best BM, Ma JD. Observations of urinary oxycodone and metabolite distributions in pain patients. J Anal Toxicol 2014;38:129–34.10.1093/jat/bku007Search in Google Scholar

6. Marsousi N, Daali Y, Rudaz S, Almond L, Humphries H, Desmeules J, et al. Prediction of metabolic interactions with oxycodone via CYP2D6 and CYP3A inhibition using a physiologically based pharmacokinetic model. CPT Pharmacometrics Syst Pharmacol 2014;3:e152.10.1038/psp.2014.49Search in Google Scholar

7. Wu LT, Pilowsky DJ, Patkar AA. Non-prescribed use of pain relievers among adolescents in the United States. Drug Alcohol Depend 2008;94:1–11.10.1016/j.drugalcdep.2007.09.023Search in Google Scholar

8. Smith MT, Watt JA, Mapp GP, Cramond T. Quantitation of oxycodone in human plasma using high-performance liquid chromatography with electrochemical detection. Ther Drug Monit 1991;13:126–30.10.1097/00007691-199103000-00007Search in Google Scholar

9. Wright AW, Lawrence JA, Iu M, Cramond T, Smith MT. Solid-phase extraction method with high-performance liquid chromatography and electrochemical detection for the quantitative analysis of oxycodone in human plasma. J Chromatogr B Biomed Sci Appl 1998;712:169–75.10.1016/S0378-4347(98)00146-7Search in Google Scholar

10. Menelaou A, Hutchinson MR, Quinn I, Christensen A, Somogyi AA. Quantification of the O- and N-demethylated metabolites of hydrocodone and oxycodone in human liver microsomes using liquid chromatography with ultraviolet absorbance detection. J Chromatogr B Analyt Technol Biomed Life Sci 2003;785:81–8.10.1016/S1570-0232(02)00856-5Search in Google Scholar

11. Smith ML, Hughes RO, Levine B, Dickerson S, Darwin WD, Cone EJ. Forensic drug testing for opiates. VI. Urine testing for hydromorphone, hydrocodone, oxymorphone, and oxycodone with commercial opiate immunoassays and gas chromatography-mass spectrometry. J Anal Toxicol 1995;19:18–26.10.1093/jat/19.1.18Search in Google Scholar

12. McKinley S, Snyder JJ, Welsh E, Kazarian CM, Jamerson MH, Klette KL. Rapid quantification of urinary oxycodone and oxymorphone using fast gas chromatography-mass spectrometry. J Anal Toxicol 2007;31:434–41.10.1093/jat/31.8.434Search in Google Scholar

13. Nowatzke W, Zeng J, Saunders A, Bohrer A, Koenig J, Turk J. Distinction among eight opiate drugs in urine by gas chromatography-mass spectrometry. J Pharm Biomed Anal 1999;20:815–28.10.1016/S0731-7085(99)00086-2Search in Google Scholar

14. Ropero-Miller JD, Lambing MK, Winecker RE. Simultaneous quantitation of opioids in blood by GC-EI-MS analysis following deproteination, detautomerization of keto analytes, solid-phase extraction, and trimethylsilyl derivatization. J Anal Toxicol 2002;26:524–8.10.1093/jat/26.7.524Search in Google Scholar PubMed

15. Wagner M, Bourgogne E, Varesio E, Hopfgartner G. Quantitation of polar analytes using column-switching: application to oxycodone and three metabolites in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2010;878:637–44.10.1016/j.jchromb.2010.01.014Search in Google Scholar PubMed

16. Dawson M, Fryirs B, Kelly T, Keegan J, Mather LE. A rapid and sensitive high-performance liquid chromatography-electrospray ionization-triple quadrupole mass spectrometry method for the quantitation of oxycodone in human plasma. J Chromatogr Sci 2002;40:40–4.10.1093/chromsci/40.1.40Search in Google Scholar PubMed

17. Neuvonen M, Neuvonen PJ. Determination of oxycodone, noroxycodone, oxymorphone, and noroxymorphone in human plasma by liquid chromatography-electrospray-tandem mass spectrometry. Ther Drug Monit 2008;30:333–40.10.1097/FTD.0b013e31816e2d4bSearch in Google Scholar PubMed

18. Fang WB, Lofwall MR, Walsh SL, Moody DE. Determination of oxycodone, noroxycodone and oxymorphone by high-performance liquid chromatography-electrospray ionization-tandem mass spectrometry in human matrices: in vivo and in vitro applications. J Anal Toxicol 2013;37:337–44.10.1093/jat/bkt042Search in Google Scholar PubMed PubMed Central

19. Gaudette F, Sirhan-Daneau A, St-Onge M, Turgeon J, Michaud V. Development of a sensitive method for the determination of oxycodone and its major metabolites noroxycodone and oxymorphone in human plasma by liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2016;1008:174–80.10.1016/j.jchromb.2015.11.035Search in Google Scholar PubMed

20. Edwards SR, Smith MT. Simultaneous determination of morphine, oxycodone, morphine-3-glucuronide, and noroxycodone concentrations in rat serum by high performance liquid chromatography-electrospray ionization-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2005;814:241–9.10.1016/j.jchromb.2004.10.035Search in Google Scholar PubMed

21. Musshoff F, Trafkowski J, Kuepper U, Madea B. An automated and fully validated LC-MS/MS procedure for the simultaneous determination of 11 opioids used in palliative care, with 5 of their metabolites. J Mass Spectrom 2006;41:633–40.10.1002/jms.1021Search in Google Scholar PubMed

22. Coles R, Kushnir MM, Nelson GJ, McMillin GA, Urry FM. Simultaneous determination of codeine, morphine, hydrocodone, hydromorphone, oxycodone, and 6-acetylmorphine in urine, serum, plasma, whole blood, and meconium by LC-MS-MS. J Anal Toxicol 2007;31:1–14.10.1093/jat/31.1.1Search in Google Scholar PubMed

23. Marchei E, Escuder D, Pallas CR, Garcia-Algar O, Gomez A, Friguls B, et al. Simultaneous analysis of frequently used licit and illicit psychoactive drugs in breast milk by liquid chromatography tandem mass spectrometry. J Pharm Biomed Anal 2011;55:309–16.10.1016/j.jpba.2011.01.028Search in Google Scholar PubMed

24. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Strategies for the assessment of matrix effect in quantitative bioanalytical methods based on HPLC-MS/MS. Anal Chem 2003;75:3019–30.10.1021/ac020361sSearch in Google Scholar PubMed

25. Ogle A, Moore K, Barrett B, Young MS, Pearson J. Clinical history and characteristics of persons with oxycodone-related deaths in Hillsborough County, Florida in 2009. Forensic Sci Int 2012;223:47–52.10.1016/j.forsciint.2012.07.016Search in Google Scholar PubMed

26. Pilgrim JL, Yafistham SP, Gaya S, Saar E, Drummer OH. An update on oxycodone: lessons for death investigators in Australia. Forensic Sci Med Pathol 2015;11:3–12.10.1007/s12024-014-9624-xSearch in Google Scholar PubMed

27. Al-Asmari AI, Anderson RA, Cooper GA. Oxycodone- related fatalities in the west of Scotland. J Anal Toxicol 2009;33:423–32.10.1093/jat/33.8.423Search in Google Scholar PubMed

28. Jannetto PJ, Wong SH, Gock SB, Laleli-Sahin E, Schur BC, Jentzen JM. Pharmacogenomics as molecular autopsy for postmortem forensic toxicology: genotyping cytochrome P450 2D6 for oxycodone cases. J Anal Toxicol 2002;26:438–47.10.1093/jat/26.7.438Search in Google Scholar PubMed

29. Visconti AJ, Santos GM, Lemos NP, Burke C, Coffin PO. Opioid overdose deaths in the city and county of San Francisco: prevalence, distribution, and disparities. J Urban Health 2015;92:758–72.10.1007/s11524-015-9967-ySearch in Google Scholar PubMed PubMed Central

30. Al-Asmari AI, Anderson RA. Method for quantification of opioids and their metabolites in autopsy blood by liquid chromatography-tandem mass spectrometry. J Anal Toxicol 2007;31:394–408.10.1093/jat/31.7.394Search in Google Scholar PubMed

31. Wolf BC, Lavezzi WA, Sullivan LM, Flannagan LM. One hundred seventy two deaths involving the use of oxycodone in Palm Beach County. J Forensic Sci 2005;50:192–5.10.1520/JFS2004194Search in Google Scholar

32. Drummer OH, Syrjanen ML, Phelan M, Cordner SM. A study of deaths involving oxycodone. J Forensic Sci 1994;39:1069–75.10.1520/JFS13685JSearch in Google Scholar

33. Schulz M, Iwersen-Bergmann S, Andresen H, Schmoldt A. Therapeutic and toxic blood concentrations of nearly 1,000 drugs and other xenobiotics. Crit Care 2012;16:R136.10.1186/cc11441Search in Google Scholar PubMed PubMed Central

34. Liukas A, Kuusniemi K, Aantaa R, Virolainen P, Neuvonen M, Neuvonen PJ, et al. Plasma concentrations of oral oxycodone are greatly increased in the elderly. Clin Pharmacol Ther 2008;84:462–7.10.1038/clpt.2008.64Search in Google Scholar PubMed

Received: 2016-10-31
Accepted: 2016-11-28
Published Online: 2017-1-12
Published in Print: 2017-8-28

©2017 Walter de Gruyter GmbH, Berlin/Boston

Articles in the same Issue

  1. Frontmatter
  2. Editorial
  3. Mass spectrometry or immunoassay: est modus in rebus
  4. Reviews
  5. The use of liquid chromatography-tandem mass spectrometry for therapeutic drug monitoring of antibiotics in cancer patients
  6. Tackling serum folate test in European countries within the health technology assessment paradigm: request appropriateness, assays and health outcomes
  7. Genetics and Molecular Diagnostics
  8. Genetic diagnosis of α1-antitrypsin deficiency using DNA from buccal swab and serum samples
  9. General Clinical Chemistry and Laboratory Medicine
  10. Serum triglyceride measurements: the commutability of reference materials and the accuracy of results
  11. Variant peptide detection utilizing mass spectrometry: laying the foundations for proteogenomic identification and validation
  12. Evaluation of two fully automated immunoassay based tests for the measurement of 1α,25-dihydroxyvitamin D in human serum and comparison with LC-MS/MS
  13. Parallel diurnal fluctuation of testosterone, androstenedione, dehydroepiandrosterone and 17OHprogesterone as assessed in serum and saliva: validation of a novel liquid chromatography-tandem mass spectrometry method for salivary steroid profiling
  14. Determination of oxycodone and its major metabolites noroxycodone and oxymorphone by ultra-high-performance liquid chromatography tandem mass spectrometry in plasma and urine: application to real cases
  15. Identification and quantitation of phosphatidylethanols in oral fluid by liquid chromatography-tandem mass spectrometry
  16. Relationship between plasma and salivary melatonin and cortisol investigated by LC-MS/MS
  17. Paramagnetic micro-particles as a tool for rapid quantification of apixaban, dabigatran, edoxaban and rivaroxaban in human plasma by UHPLC-MS/MS
  18. Measurements of serum non-ceruloplasmin copper by a direct fluorescent method specific to Cu(II)
  19. The serum concentrations of leptin and MCP-1 independently predict low back pain duration
  20. Immunoassay screening in urine for synthetic cannabinoids – an evaluation of the diagnostic efficiency
  21. Cancer Diagnostics
  22. Study of kallikrein-related peptidase 6 (KLK6) and its complex with α1-antitrypsin in biological fluids
  23. Cardiovascular Diseases
  24. A candidate liquid chromatography mass spectrometry reference method for the quantification of the cardiac marker 1-32 B-type natriuretic peptide
  25. The natriuretic peptide MR-proANP predicts all-cause mortality and adverse outcome in community patients: a 10-year follow-up study
  26. CASZ1 loss-of-function mutation contributes to familial dilated cardiomyopathy
  27. Diabetes
  28. Evaluating new HbA1c methods for adoption by the IFCC and NGSP reference networks using international quality targets
  29. Infectious Diseases
  30. Analytical and diagnostic performance of two automated fecal calprotectin immunoassays for detection of inflammatory bowel disease
  31. Letters to the Editor
  32. Is fasting necessary for lipid profile determinations? Some considerations from the perspective of the clinical laboratory
  33. Precision of nonfasting lipid profiles should focus on clinical relevance rather than necessarily obtaining the least variation
  34. Triglyceride concentrations should be measured after elimination of free glycerol to exclude interindividual variations due to adiposity and fasting status
  35. Estimation of the reference interval for serum folate measured with assays traceable to the WHO International Standard
  36. Implausible elevation of peripheral thyroid hormones during therapy with a protein supplement
  37. Interference in Na+ measurements on the Siemens RAPIDPoint® 500 after nortriptyline intoxication: a case report
  38. Usefulness of maternal red cell antibodies to predict hemolytic disease of the fetus and newborn and significant neonatal hyperbilirubinemia: a retrospective study
  39. Improvement of the Sandell-Kolthoff reaction method (ammonium persulfate digestion) for the determination of iodine in urine samples
  40. Clinical use of targeted high-throughput whole-genome sequencing for a dengue virus variant
Downloaded on 8.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/cclm-2016-0990/html?lang=en&srsltid=AfmBOopx-m_w2QDhkzODA8sWIcbXhei6J-MmFAysmQEdZqvVMRbDEDyi
Scroll to top button