Skip to main content
Cannabis and Cannabinoid Research logoLink to Cannabis and Cannabinoid Research
. 2023 Aug 9;8(4):657–669. doi: 10.1089/can.2021.0181

Simultaneous Assessment of Serum Levels and Pharmacologic Effects of Cannabinoids on Endocannabinoids and N-Acylethanolamines by Liquid Chromatography–Tandem Mass Spectrometry

Timothy A Couttas 1,, Carola Boost 2,, Franziska Pahlisch 2, Eliska B Sykorova 1,2, Judith E Leweke 1,3, Dagmar Koethe 1,2, Heike Endepols 4,5,6,7, Cathrin Rohleder 1,2,3,4,8,, F Markus Leweke 1,2,*,
PMCID: PMC10442685  PMID: 35580134

Abstract

Introduction:

The primary compounds of Cannabis sativa, delta-9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD), inflict a direct influence on the endocannabinoid system-a complex lipid signaling network with a central role in neurotransmission and control of inhibitory and excitatory synapses. These phytocannabinoids often interact with endogenously produced endocannabinoids (eCBs), as well as their structurally related N-acylethanolamines (NAEs), to drive neurobiological, nociceptive, and inflammatory responses. Identifying and quantifying changes in these lipid neuromodulators can be challenging owing to their low abundance in complex matrices.

Materials and Methods:

This article describes a robust liquid chromatography–tandem mass spectrometry (LC-MS/MS) method for the extraction and quantification of the eCBs anandamide and 2-arachidonoylglycerol, along with their congener NAEs oleoylethanolamine and palmitoylethanolamine, and phytocannabinoids CBD, Δ9-THC, and 11-Nor-9-carboxy-Δ9-tetrahydrocannabinol, a major metabolite of Δ9-THC. Our method was applied to explore pharmacokinetic and pharmacodynamic effects from intraperitoneal injections of Δ9-THC and CBD on circulating levels of eCBs and NAEs in rodent serum.

Results:

Detection limits ranged from low nanomolar to picomolar in concentration for eCBs (0.012–0.24 pmol/mL), NAEs (0.059 pmol/mL), and phytocannabinoids (0.24–0.73 pmol/mL). Our method displayed good linearity for calibration curves of all analytes (R2>0.99) as well as acceptable accuracy and precision, with quality controls not deviating >15% from their nominal value. Our LC-MS/MS method reliably identified changes to these endogenous lipid mediators that followed a causal relationship, which was dependent on both the type of phytocannabinoid administered and its pharmaceutical preparation.

Conclusion:

We present a rapid and reliable method for the simultaneous quantification of phytocannabinoids, eCBs, and NAEs in serum using LC-MS/MS. The accuracy and sensitivity of our assay infer it can routinely monitor endogenous levels of these lipid neuromodulators in serum and their response to external stimuli, including cannabimimetic agents.

Keywords: cannabidiol, endocannabinoids, liquid chromatography–tandem mass spectrometry, N-acylethanolamines, rat serum, Δ9-tetrahydrocannabinol

Introduction

Endocannabinoids (eCBs) and their structurally related N-acylethanolamines (NAEs) are endogenous lipid mediators widely distributed throughout the central nervous system (CNS) and periphery.1 Anandamide (AEA) and 2-arachidonoylglycerol (2-AG) are the primary eCB agonists of the endocannabinoid system (ECS), a key homeostatic system involved in neurotransmission and several regulatory processes.2 AEA and 2-AG act through at least two subtypes of G protein-coupled cannabinoid receptors: cannabinoid receptor 1 (CB1R) and cannabinoid receptor 2 (CB2R), which are primarily expressed in various brain regions and immune cells, respectively.3,4

Alterations to these eCBs have been found in several neuropsychiatric conditions, including schizophrenia,5,6 borderline personality and post-traumatic stress disorders,7 Alzheimer's,8,9 and Parkinson's disease.10 The NAEs oleoylethanolamine (OEA) and palmitoylethanolamine (PEA) exert their influence on various physiological functions, including neuroprotection, inflammation, and satiety.11–13

Although structurally analogous to AEA, NAE receptor signaling does not occur through CB1/2R but involves isoforms of the peroxisome proliferator-activated receptor family, transient receptor potential cation channel subfamily V member 1, or G protein-coupled receptor 55.14–17 OEA and PEA may indirectly impact CB1/2R-mediated signaling through their shared biosynthesis and degradation with AEA,18 having demonstrated the capacity to suppress AEA degradation through direct competition19,20 and decreased expression21 of fatty acid amide hydrolase (FAAH), their primary catabolic enzyme.22 The AEA-NAE interplay and the ubiquity of the ECS have led to increased interest of their profiles in physiology, neurology, and their response to cannabis constituents.

Exposure to delta-9-tetrahydrocannabinol (Δ9-THC), the principal psychoactive component of cannabis,23 results in overactivation of the ECS, with chronic exposure during adolescence resulting in long-lasting, potentially irreversible neurobiological alterations in various brain regions.24 Frequent cannabis use reduces cerebrospinal fluid AEA levels in schizophrenia and is considered a risk factor for disease development.25 In rodents, downregulation of AEA signaling occurs in the CNS following recurrent intraperitoneal (i.p.) injections of Δ9-THC.26 Reductions in locomotor activity and prepulse inhibition of the acoustic startle response have also been observed following acute Δ9-THC administration; however, the effects were dependent on the pharmacokinetic properties of the delivery solvent.27

Clinical trials administering the main nonpsychotomimetic compound in cannabis, cannabidiol (CBD), yield improvements in psychotic episodes without adverse side effects.28,29 Clinical improvement is accompanied by an increase in AEA and congener OEA and PEA ligands.28 This suggests that CBDs antipsychotic properties are mediated through FAAH inhibition or blockade of fatty acid-binding proteins (FABPs), which act as intracellular carriers.30,31 The lipophilic nature of these eCBs and NAEs also allows them to modulate and readily cross the blood–brain barrier,32–34 making them promising biomarker candidates and therapeutic targets.35,36

Quantification of eCBs and NAEs relies on mass spectrometry as their concentrations are often found at trace levels under physiological conditions, making their detection difficult.37 Liquid chromatography–tandem mass spectrometry (LC-MS/MS) with collision-induced dissociation (CID) is conventional for their analysis, as the additional structural information obtained from CID fragment ions can be used for selective reaction monitoring and multiple reaction monitoring (MRM) to improve sensitivity and reduce background interference.38 LC-MS/MS methods have been developed for eCBs,38–42 NAEs,40–45 and phytocannabinoids46–49 in various biological tissues. However, to the best of our knowledge, no single LC-MS/MS method has been established that monitors both endogenous and exogenous cannabinoids.

In this study we present a robust LC-MS/MS method with the capacity to reliably identify and quantify common eCBs, NAEs, and phytocannabinoids in a single assay. We successfully used our LC-MS/MS method in serum from a pharmacokinetics study on rats given i.p. injections of Δ9-THC or CBD to examine their associated effects on endogenous eCB and NAE lipid mediators and evaluate differential effects between pharmaceutical preparations.

Materials and Methods

Reagents and standards

Standards for eCBs and NAEs, including their deuterated counterparts, were purchased from Cayman Chemical (United States). Purified natural CBD and [2H3]-CBD were supplied by THC Pharm GmbH (Germany). Standards for Δ9-THC, 11-Nor-9-carboxy-Δ9-tetrahydrocannabinol (Δ9-THC-COOH), [2H3]-Δ9-THC, and [2H3]-Δ9-THC-COOH were provided by Lipomed AG (Switzerland). Solvents for lipid extraction and LC-MS/MS were purchased from Honeywell Specialty Chemicals (Germany).

Calibration curves and quality controls

Twelve-point calibration curves were prepared by serial dilutions in methanol, over a concentration range of 0.012–25 pmol/mL for AEA, 0.049–100 pmol/mL for OEA/PEA, 0.244–500 pmol/mL for 2-AG, 0.122–250 pmol/mL for Δ9-THC/CBD, and 0.73–1500 pmol/mL for Δ9-THC-COOH. Calibrators were spiked with an internal standard (IS) cocktail, comprising [2 H4]-AEA (25 pmol), [2H2]-OEA and [2 H4]-PEA (100 pmol), [2 H8]-2-AG (500 pmol), [2 H3]-Δ9-THC (100 pmol), [2 H3]-Δ9-THC-COOH, and [2 H3]-CBD (250 pmol). Peak areas for each analyte were normalized against their respective deuterated IS.

Quality controls (QCs) were generated from independently prepared stock solutions at four concentrations for AEA (12.5, 1.56, 0.39, and 0.049 pmol/mL), OEA/PEA (50, 6.25, 1.56, and 0.195 pmol/mL), 2-AG (125, 31.25, 7.81, and 0.98 pmol/mL), CBD/Δ9-THC (250, 62.5, 15.63, and 1.95 pmol/mL), and Δ9-THC-COOH (750, 93.75, 23.44, and 2.93 pmol/mL). These concentrations represent the high (Q1), middle (Q2), and low (Q3) range of the calibration curves, and the lower limits of quantification (LLOQ; Q4). QCs were loaded with the same IS.

Serum and sample extraction

Control human serum was obtained from our prior schizophrenia-related study,25 with rodent serum (n=28) obtained from investigations into behavioral changes following i.p. administration with Δ9-THC (5 mg/kg, n=14) or CBD (12 mg/kg, n=14) prepared in ethanol:Tween 80:saline (aqueous; with ethanol as a cosolvent and Tween-80 as surfactant, 1:1:18; n=8 for Δ9-THC, n=6 for CBD) or sesame oil (lipid; n=6 for Δ9-THC, n=8 for CBD). The Ethics Committees of the Medical Faculty Cologne, University of Cologne, Germany (00-053) and the Medical Faculty Mannheim, Heidelberg University, Germany (2009-235N-MA) approved the use of human serum samples for this research. The animal study was approved by the regional authority State Agency for Nature, Environment and Consumer Protection of the State North Rhine-Westphalia (LANIUV-NRW).

Rodent blood was withdrawn from the femoral artery 120 min postinjection and subsequently centrifuged (2054 g, 4°C) for 30 min. Serum aliquots (1 mL) were stored at −80°C until extraction. Serum aliquots were spiked with IS and extracted under chloroform/methanol (2:1, v/v).43 Extractions were performed at low temperatures (<4°C) to avoid artefactual AEA formation.50 Samples were dried under N2, reconstituted in methanol (80 μL), and transferred to MS vials (Brown Chromatography Supplies GmbH, Germany).

Quantification by LC-MS/MS

LC-MS/MS was performed using an API 5000 triple quadrupole mass spectrometer (Sciex), coupled to an Agilent 1200 HPLC system (Agilent Technologies). Samples (20 μL) were injected using a CTC PAL Autosampler set at 4°C (CTC Analytics AG, Switzerland). Analytes were resolved through chromatographic separation using a 4-μm Synergi Hydro-RP C18 column (150×2 mm; Phenomenex, Torrance, CA), with column chamber set at 40°C, over a binary gradient with a flow rate of 0.5 mL/min. HPLC gradient conditions were as follows: 0 min, 25:75 A/B; 2.5 min, 20:80 A/B; 7.5 min, 10:90 A/B; 8 min, 0:100 A/B; 10 min, 25:75 A/B; 18 min, 25:75 A/B. Solvent A: 0.1% formic acid in water; Solvent B: methanol. Total run time was 18 min.

LC-MS/MS was performed in positive ion mode, [M + H]+, with quantifier and qualifier ion transitions selected for each analyte, at a dwell time of 50 ms. Source parameters were set as follows: positive ion spray voltage, 5000 V; ion source temperature, 500°C; collision gas, 7 psi; curtain gas, 35 psi; nebulizer gas, 25 psi; turbo gas, 45 psi. Transitions were optimized using direct infusion (10 μL/min) with each standard (100 ng/mL). MS/MS parameters are summarized in Table 1. Data were acquired and processed using Analyst® (Sciex), version 1.6.2.

Table 1.

Operational Parameters for Liquid Chromatography–Tandem Mass Spectrometry

Analyte Precursor→product (m/z) DP [V] CE [V] CXP [V]
AEA 348.3→62.0 21 25 24
348.3→44.0 21 67 18
[2H4]-AEA 352.4→65.9 71 27 12
352.4→48.3 71 65 10
OEA 326.4→62.0 211 27 12
326.4→44.0 211 61 20
[2H2]-OEA 328.4→62.0 136 71 10
328.4→44.0 136 89 18
PEA 300.3→62.0 36 23 24
300.3→43.9 36 53 18
[2H4]-PEA 304.4→61.9 86 23 14
304.4→44.0 86 63 18
2-AG 379.3→287.2 86 17 40
379.3→90.9 86 67 24
[2H8]-2-AG 387.3→294.2 161 17 35
387.3→91.0 161 68 20
CBD/Δ9-THC 315.3→193.1 96 31 20
315.3→41.0 96 85 10
[2H3]-CBD/[2H3]-Δ9-THC 318.3→196.1 101 31 20
318.3→41.0 101 85 10
Δ9-THC-COOH 345.3→327.3 176 23 14
345.3→299.3 176 29 22
[2H3]-Δ9-THC-COOH 348.3→330.3 16 21 40
348.3→302.3 16 29 42

Transitions in italics were used as qualifier ions.

Δ9-THC, delta-9-tetrahydrocannabinol; Δ9-THC-COOH, 11-Nor-9-carboxy-Δ9-tetrahydrocannabinol; 2-AG, 2-arachidonoylglycerol; AEA, anandamide; CBD, cannabidiol; CE, collision energy; CXP, collision cell exit potential; DP, declustering potential; m/z, mass to charge ratio; OEA, oleoylethanolamine; PEA, palmitoylethanolamine.

Method validation

Our LC-MS/MS method was validated in accordance with the international requirements and regulatory guidelines for the validation of quantitative methods.51–54 Analyte specificity, calibration curve linearity, sensitivity, limit of detection (LOD) and LLOQ, intra- and interday accuracy, sample recovery, matrix effect, precision, and stability were assessed. Extended details on validation measures are provided (Supplementary Data).

Statistical analysis

Serum concentrations of Δ9-THC, Δ9-THC-COOH, and CBD were compared between aqueous and lipid formulations using two-tailed, unpaired t-tests, corrected for multiple comparisons using the Holm–Šídák method (GraphPad Prism, version 9.1.0). Phytocannabinoid correlations with eCBs and NAEs were log-transformed (natural log) and analyzed by Pearson analysis. Grubb's test at a high stringency (Q=1%) was used to identify and remove a single statistical outlier for 2-AG following i.p. administered CBD with aqueous delivery formulation.

Linear regression analysis was applied to identify interaction effects between phytocannabinoid associations with eCBs and/or NAEs, using solvent delivery as the response variable, and adjusted for multiple comparisons using Bonferroni. For all experiments, statistical significance was established at p<0.05.

Results

LC-MS/MS specificity

Direct infusion of standards allowed for CID fragmentation and characterization of [M + H]+ ions, which were used for LC-MS/MS in MRM mode. The strongest fragment ions were selected for quantification, with the second selected as a qualifier transition. Chemical structures and fragmentation patterns of analytes are given in Figure 1. Points of fragmentation for the resultant quantifier (red) and qualifier (blue) ions are also presented. CID parameters for transitions, including IS, are given in Table 1. Quantifier/qualifier ion ratios remained within ±25% range of our reference values (data not shown). All analytes yielded unique precursor–product ion pairs, except Δ9-THC/CBD, which produced the same ion transitions (Table 1; Fig. 1). Their identification was resolved through chromatographic separation (Fig. 2).

FIG. 1.

FIG. 1.

Chemical structures and MS/MS fragmentation characteristics following CID for (A) AEA, (B) 2-AG, (C) OEA, (D) PEA, (E) CBD, (F) Δ9-THC, and (G) Δ9-THC-COOH. Structurally diagnostic product ions used for quantitation (red) and qualifier ions (blue) have been displayed along with their expected m/z values. Δ9-THC, delta-9-tetrahydrocannabinol; Δ9-THC-COOH, 11-Nor-9-carboxy-Δ9-tetrahydrocannabinol; 2-AG, 2-arachidonoylglycerol; AEA, anandamide; CBD, cannabidiol; CID, collision-induced dissociation; m/z, mass to charge ratio; MS/MS, tandem mass spectrometry; OEA, oleoylethanolamine; PEA, palmitoylethanolamine.

FIG. 2.

FIG. 2.

Observed chromatograms for (A) naturally occurring and (B) deuterated eCBs, NAEs, and phytocannabinoids. Peak intensity of analytes, in cps, is plotted against retention time in minutes. Resolution of isobaric Δ9-THC and CBD species was achieved using LC separation with a C18 column and a methanol-water gradient. cps, counts per second; eCBs, endocannabinoids; LC, liquid chromatography; NAEs, N-acylethanolamines.

LC conditions allowed for appropriate separation of all analytes, with desirable peak shape and signal intensity to perform accurate quantification (Fig. 2). AEA, 2-AG, OEA, PEA, and Δ9-THC-COOH were resolved at retention times (RTs) 6.50±0.01, 6.73±0.02, 7.65±0.01, 7.20±0.01, and 4.03±0.02 min, respectively, each with a relative standard deviation of <3%. Isobaric compounds CBD and Δ9-THC were resolved at 4.28±0.02 and 6.54±0.02 min, respectively (Fig. 2). The RTs of all IS were comparable with their naturally occurring counterparts (Fig. 2). No peaks were observed for IS in nonspiked samples (data not shown).

Instrument sensitivity and linearity

Sensitivity of analytes was measured with respect to the LOD and LLOQ. LOD for AEA and 2-AG was 0.012 and 0.24 pmol/mL, respectively, with OEA and PEA both achieving 0.059 pmol/mL. CBD, Δ9-THC, and Δ9-THC-COOH LOD were 0.24, 0.49, and 0.73 pmol/mL, respectively. LLOQ was 0.049 for AEA, 0.195 for OEA and PEA, 0.98 for 2-AG, 1.95 for CBD and Δ9-THC, and 2.93 pmol/mL for Δ9-THC-COOH. Representative chromatograms from LLOQ have been provided (Supplementary Fig. S1).

Calibration curves (n=3) plotting the quotient peak area of analytes (normalized to their IS), against concentration, were constructed using a weighted linear regression analysis (w=1/x). Linearity was observed over the concentration range of 0.049–25 pmol/mL for AEA (R2=0.9973), 0.195–100 pmol/mL for OEA (R2=0.9990) and PEA (R2=0.9985), 0.98–250 pmol/mL for 2-AG (R2=0.9999), 1.95–250 pmol/mL for CBD (R2=0.9952) and Δ9-THC (R2=0.9982), and 2.93–1500 pmol/mL for Δ9-THC-COOH (R2=0.9901).

Accuracy and precision

Accuracy and precision were measured across a single run (intraday) and three consecutive days (interday) through QC analyses (n=3). Our results show that the precision and accuracy for all analytes assayed were within our acceptance criteria, with QCs not deviating >15% (20% for the LLOQ) of the expected value (Table 2).

Table 2.

Intraday and Interday Accuracy and Precision of Quality Controls

Analyte QC level Exp Conc (pmol/mL) Intraday
Interday
Avg Conc (pmol/mL) SD Accuracy (RE, %) Precision (RSD, %) Avg Conc (pmol/mL) SD Accuracy (RE, %) Precision (RSD, %)
AEA QC1 12.50 14.07 0.47 12.5 3.4 14.17 0.38 13.4 2.7
  QC2 1.56 1.50 0.08 −3.8 5.3 1.60 0.07 2.6 4.4
QC3 0.39 0.35 0.01 −9.7 1.5 0.38 0.04 −2.6 10.5
QC4 0.05 0.05 0.00 −4.1 7.7 0.04 0.00 −9.2 9.0
2-AG QC1 250.00 285.33 1.53 14.1 0.5 280.00 8.72 12.0 3.1
  QC2 31.25 34.43 0.06 10.2 0.2 33.30 1.10 6.6 3.3
QC3 7.81 8.06 0.87 3.2 10.7 8.11 0.91 3.8 11.2
QC4 0.98 1.09 0.07 11.2 6.5 1.09 0.08 11.2 7.0
OEA QC1 50.00 49.70 3.14 −0.6 6.3 49.07 2.86 −1.9 5.8
  QC2 6.25 6.02 0.61 −3.7 10.0 5.63 0.27 −9.9 4.7
QC3 1.56 1.39 0.03 −10.9 2.1 1.37 0.06 −12.2 4.1
QC4 0.20 0.19 0.02 −2.6 8.9 0.20 0.02 2.6 11.5
PEA QC1 50.00 43.51 3.30 −13.0 7.6 44.83 2.05 −10.3 4.6
  QC2 6.25 5.43 0.17 −13.1 3.2 5.34 0.25 −14.6 4.6
QC3 1.56 1.41 0.14 −9.6 9.6 1.46 0.09 −6.4 6.3
QC4 0.20 0.20 0.02 2.6 8.5 0.21 0.01 7.7 5.7
CBD QC1 250.00 260.33 18.58 4.1 7.1 254.33 30.62 1.7 12.0
  QC2 31.25 28.33 1.29 −9.3 4.5 27.77 1.55 −11.1 5.6
QC3 7.81 7.09 0.28 −9.2 3.9 7.11 0.84 −9.0 11.8
QC4 0.98 0.94 0.08 −4.1 8.1 0.95 0.05 −3.1 5.3
Δ9-THC QC1 125.00 120.00 3.46 −4.0 2.9 130.00 12.29 4.0 9.5
  QC2 31.25 27.23 2.84 −12.9 10.4 29.77 0.85 −4.7 2.9
QC3 7.81 7.81 0.76 0.0 9.7 7.88 0.80 0.9 10.2
QC4 1.95 1.82 0.36 −6.7 19.7 2.19 0.18 12.3 8.0
Δ9-THC-COOH QC1 750.00 759.67 109.44 1.3 14.4 848.00 106.10 13.1 12.5
  QC2 93.75 105.33 3.51 12.4 3.3 105.00 3.46 12.0 3.3
QC3 23.44 22.30 2.80 −4.9 12.6 25.37 0.92 8.2 3.6
QC4 2.93 3.39 0.10 15.7 2.8 2.94 0.49 0.3 16.7

Accuracy was reported as percentage RE for the measured mean of spiked QCs against the nominal target value. Precision was calculated as the percentage of RSD from repeated QC measurements.

Avg Conc, average concentration from repeated QC measurements (n=3); Exp Conc, expected concentration; QC, quality control; RE, relative error; RSD, relative standard deviation; SD, standard deviation.

Stability

Room temperature and 4°C were assessed by reanalyzing QCs, 24 h after initial screening. Freeze-thaw stability was measured following three cycles of QC thawing from storage conditions (24 h at −20°C). Long-term effects were examined on QCs held at −20°C for 3 months, compared with freshly prepared stocks. Analytes were predominately stable across QCs, with <15% variance from the nominal value (Table 3). Exceptions to our acceptance criteria included OEA (−15.9%), 2-AG (39.5%), and Δ9-THC (−18.6%) at room temperature, CBD (23.2%) and Δ9-THC-COOH (−33.7%) held at 4°C for 24 h. All analytes were within permissible variability following 3 months of storage at −20°C. However, inconsistencies to PEA (25.4%) and CBD (−24.2%) were observed after multiple freeze-thaw cycles.

Table 3.

Stability Under Experimental and Storage Conditions

Analyte Room temp
4°C
Freeze–thaw
Long-term
QC1 QC2 QC3 QC1 QC2 QC3 QC1 QC2 QC3 QC1 QC2 QC3
AEA −7.5 3.5 −10.3 2.2 −10.9 −3.9 0.7 2.6 −2.6 1.8 −5.2 7.7
2-AG 1.8 −5.5 39.5 −1.8 −6.1 −13.5 0.0 1.7 −3.0 14.3 5.4 4.8
OEA 9.1 −15.9 8.0 −11.0 6.9 −2.2 −1.0 −4.2 2.8 −0.3 −2.7 5.9
PEA 5.0 −0.2 3.0 −8.7 −4.4 −5.1 −6.0 2.8 25.4 2.1 12.4 14.4
CBD −7.9 −2.9 −7.5 10.0 12.8 23.2 −24.2 −11.5 13.4 14.6 1.9 −14.9
Δ9-THC −18.6 3.6 −10.2 −3.3 5.3 5.8 −3.3 −0.8 −10.7 12.7 8.6 4.5
Δ9-THC-COOH 7.5 −1.8 10.3 −3.2 −33.7 6.3 −5.3 −3.8 11.9 13.6 8.5 9.3

Data are expressed as mean deviation (%) from initial reference. Deviations considered significant are illustrated in bold (>±15%).

Matrix effects and sample recovery

Sample recovery and matrix effects were assessed in human serum, collected from the same healthy volunteer. Although eCBs and NAEs have been readily detected in both plasma and serum, evidence suggests their measurement is more reliable in serum owing to higher concentrations,55,56 which was the basis for its selection. Spiked replicates (n=3) were prepared for each condition (pre-extraction, postextraction, and neat methanol) using QC1, 2, and 3 concentrations as reference. Endogenous levels of eCBs and NAEs in serum were also analyzed (blank matrix, n=3; Supplementary Fig. S1), and their average was subtracted from spike response for a more accurate reading.

Matrix effects were deemed acceptable once normalized against their IS, with variance not exceeding 15% (Table 4). Recovery of eCBs and NAEs ranged from 87.3% to 99.8% and 76.2% to 99.7% for phytocannabinoids (Table 4). Similar yields of recovery were reported previously (82–99%) in serum43; however, we achieved between 2- to 10-fold higher sensitivity with the current method. 2-AG recovery was notably higher compared with prior investigations in whole blood (36.9–53.0%) and plasma (42.7%).39,57 Phytocannabinoid recovery corresponded to prior values for Δ9-THC and Δ9-THC-COOH (80–99%) in plasma, Δ9-THC-COOH in whole blood (73%) and exceeded previous yields of Δ9-THC (59%) and CBD (73%) extracted from whole blood, and CBD in plasma (60–70%).47–49

Table 4.

Matrix Effect and Analyte Recovery

Analyte Matrix effect
Normalized matrix effect
Sample recovery
QC1 QC2 QC3 QC1 QC2 QC3 QC1 QC2 QC3
AEA 11.4 14.8 13.6 7.1 7.9 2.9 89.8 99.8 99.1
OEA 24.0 16.2 20.9 9.2 7.1 3.7 91.1 88.9 87.3
PEA 8.7 17.6 28.2 5.4 7.6 14.9 96.4 96.6 95.3
2-AG 21.1 20.5 34.6 9.7 5.8 14.4 91.6 98.0 94.2
CBD 13.5 10.1 27.2 5.1 3.4 10.2 95.2 99.7 97.9
Δ9-THC 12.2 18.9 11.7 5.8 9.2 8.3 84.1 99.3 93.5
Δ9-THC-COOH 11.4 14.8 13.6 5.3 7.9 6.7 76.2 79.7 85.6

Values are expressed as mean % (n=3).

Monitoring eCB/NAE expression against administered phytocannabinoid concentration

Our LC-MS/MS method was used to measure eCBs and NAEs against phytocannabinoid concentrations in rat serum following i.p. injections with Δ9-THC and CBD, prepared in both aqueous and lipid-based formulations for delivery comparison. Aqueous formulation yielded significantly greater serum levels of Δ9-THC (p=0.003), and its metabolite Δ9-THC-COOH (p=0.012), than the lipid formulation 120 min postinjection, whereas no discernible differences to CBD levels were observed between delivery solvents (Fig. 3A). Solvent properties appear to have also influenced Δ9-THC associations with eCBs and NAEs, as each eCB/NAE correlation was contrary between the two formulations (Table 5).

FIG. 3.

FIG. 3.

Effects on AEA in serum following Δ9-THC and CBD i.p. administration. (A) Concentrations of Δ9-THC, Δ9-THC-COOH, and CBD in serum 120 min after i.p. injection (5 mg/kg Δ9-THC; 12 mg/kg CBD). Results show mean and standard error for different delivery solvents (aqueous and lipid). A statistically significant difference in Δ9-THC (p=0.003) and Δ9-THC-COOH (p=0.012) between aqueous and lipid solvents was identified by unpaired t-tests. (B–G) display correlations for AEA with (B, C) Δ9-THC, (D, E) Δ9-THC-COOH, and (F, G) CBD when either (B, D, F) aqueous or lipid-based (C, E, G) solvent formulations were used. A line of best fit (linear regression) is shown. Correlations were determined by Pearson analysis at a confidence interval of 95%. The coefficient of correlation (r) and p-values are given. i.p., intraperitoneal; ns, not significant.

Table 5.

Phytocannabinoid Correlations with Endocannabinoids and N-Acylethanolamines

  Analyte AEA OEA PEA 2-AG
Δ9-THC (aqueous) r −0.8432 −0.0073 −0.4161 0.4411
p 0.0085 0.9864 0.3051 0.2739
Δ9-THC (lipid) r 0.7121 0.6188 0.6164 −0.3188
p 0.1124 0.1903 0.1925 0.5381
Δ9-THC-COOH (aqueous) r −0.8088 −0.3740 −0.0041 0.4302
p 0.0151 0.3614 0.9923 0.2874
Δ9-THC-COOH (lipid) r 0.8803 0.6244 0.8657 −0.7356
p 0.0206 0.1852 0.0259 0.0956
CBD (aqueous) r 0.9043 0.7483 0.7610 0.7858
p 0.0133 0.0871 0.0789 0.0639
CBD (lipid) r 0.7079 0.7036 0.5172 −0.1244
p 0.0495 0.0515 0.1893 0.7692

Pearson correlation analysis was used to determine associations between exogenous (Δ9-THC, CBD) and endogenous (AEA, OEA, PEA, 2-AG) cannabinoids (log-transformed). Coefficients of correlation (r) and p-values are given, with significance illustrated in bold.

Linear regression verified significant opposing associations between the delivery solvents for Δ9-THC (p=0.006) and Δ9-THC-COOH (p<0.0001) with AEA (Fig. 3B–E), which were the result of significantly negative relationships with aqueous Δ9-THC administration (Δ9-THC-AEA: r=−0.84, p=0.009; Δ9-THC-COOH-AEA: r=−0.81, p=0.015) and positive associations with lipid-based delivery (Δ9-THC-AEA: r=0.71, p=0.112; Δ9-THC-COOH-AEA: r=0.88, p=0.021). CBD interactions with AEA and its congeners were consistent between delivery formulations (Fig. 3E, F; Table 5).

Pearson analysis showed a significantly positive association for CBD-AEA in both aqueous (r=0.91, p=0.013) and lipid preparations (r=0.71, p=0.049). Positive trends were also observed with OEA (aqueous: r=0.74, p=0.087; lipid: r=0.70, p=0.052) and PEA (aqueous: r=0.76, p=0.080; lipid: r=0.61, p=0.113). CBD exhibited no significant association with 2-AG when either aqueous (r=0.23, p=0.706) or lipid-based (r=−0.15, p=0.721) formulation was used.

Discussion

This study presents an LC-MS/MS method capable of accurately quantifying endogenous and exogenous cannabinoids in a single assay with comparable39,47,49 or greater sensitivity40–42,44–46,48 to prior approaches. Our method was successfully applied to monitor endogenous levels of eCBs and NAEs in human serum and showed its capacity to measure their associations in response to Δ9-THC or CBD injections in rats. Our findings support prior clinical evidence that CBD positively regulates AEA levels, along with its congeners in serum.28

Future examination is warranted to assess whether the mechanism responsible for the observed CBD-AEA/NAE trends in our data are a consequence of CBDs capacity to inhibit their shared FAAH hydrolysis and FABPs-mediated intracellular transport. Although positive correlations were also observed with OEA and PEA, the degree of association was not as significant, suggesting the underlying mechanism may be more complex than blockage of their shared degradation pathway.

Δ9-THC correlations with eCBs and NAEs were more confounding, particularly the opposing directionality with different delivery formulations. Previously, we highlighted only aqueous i.p. injections of Δ9-THC elicit behavioral abnormalities in rats owing to a faster kinetic than the lipid formulation, most likely owing to a different amount of CB1R activation per time unit.27 The relationship between the speed of drug delivery and physiological and neuropsychological effects has also been observed in studies analyzing the effects of drugs such as cocaine or methylphenidate. Thereby, the time to peak effect had been suggested to be critical for the reward effects, possibly because of rapid changes in dopamine release.58–60

We speculate a faster rate of Δ9-THC accrual with the aqueous solvent may explain its five-fold higher concentration in serum compared with the lipid formulation, and the significant inverse association observed with AEA that aligns with prior literature.25,26

Δ9-THC-COOH response corroborates our Δ9-THC findings, as this metabolite is used as an indicator of cannabis consumption owing to Δ9-THC rate of oxidation and stability of Δ9-THC-COOH,48 and its reported association with low AEA at high concentrations.61 Impact on AEA may be a consequence of Δ9-THC-CB1R-activated AEA membrane transport62 for intracellular degradation by FAAH.63 Alternatively, AEA synthesis has been described as an “on-demand” process linked to its receptor coupling.2 As Δ9-THC and AEA are both CB1R-selective agonists,22 negative feedback through competitive CB1R binding may impact AEA production. This latter mechanism may explain why similar effects on OEA and PEA were not observed.

To the best of our knowledge, this is the first reported application of LC-MS/MS that combines exogenous and endogenous cannabinoid assessment in a single assay. Given our assay can routinely measure low concentrations of these lipid mediators in serum and the versatility of the LC conditions used, our assay has the potential to be extended to other biological matrices following adaptations to the extraction procedure, if necessary.

Although our method boasts several developments, care must still be taken to avoid interference by ion suppression, especially when switching between matrices (e.g., whole blood, plasma, and brain tissue) that display different levels of recovery.39–49 2-AG quantification is additionally complicated by its spontaneous isomerization to 1-AG, particularly evident in polar solvents that have a higher degree of acyl migration. Because of inconstant levels of expression, effects of 1-AG isomerization were not examined in this study, although their separation was feasible (Supplementary Fig. S1).

Cannabinoid instability is also a risk, as degradation and adherence to plastics are common and result in poor detection and unreliable quantitation.64,65 We recommend samples to be stored at a minimum of −20°C, aliquoted to prevent freeze–thawing with multiple analyses, and assessed within 24 h to reduce the likelihood of inconsistent results. It should be noted that stability measurements were used on QCs, owing to limited sample availability, which should be acknowledged as a limitation. Nevertheless, we have previously shown the stability of these endogenous analytes in serum when extracted under the same procedure.43 Consideration should also be given to the “delivery” of cannabimimetic agents to ensure that the appropriate response is achieved.

Supplementary Material

Supplemental data
Suppl_Data.docx (14.2KB, docx)
Supplemental data
Suppl_FigureS1.docx (262.8KB, docx)

Acknowledgment

The authors thank Prof. Rudolf Graf (†) for his invaluable support.

Abbreviations Used

Δ9-THC

delta-9-tetrahydrocannabinol

Δ9-THC-COOH

11-Nor-9-carboxy-Δ9-tetrahydrocannabinol

2-AG

2-arachidonoylglycerol

AEA

anandamide

CB1R

cannabinoid receptor 1

CB2R

cannabinoid receptor 2

CBD

cannabidiol

CE

collision energy

CID

collision-induced dissociation

CNS

central nervous system

cps

counts per second

CXP

collision cell exit potential

DP

declustering potential

eCBs

endocannabinoids

ECS

endocannabinoid system

FAAH

fatty acid amide hydrolase

FABPs

fatty acid-binding proteins

i.p.

intraperitoneal

IS

internal standard

LC-MS/MS

liquid chromatography–tandem mass spectrometry

LLOQ

lower limits of quantification

LOD

limit of detection

m/z

mass to charge ratio

MRM

multiple reaction monitoring

NAEs

N-acylethanolamines

OEA

oleoylethanolamine

PEA

palmitoylethanolamine

QCs

qquality controls

RE

relative error

RSD

relative standard deviation

RTs

retention times

SD

standard deviation

Author Disclosure Statement

F.M.L. and D.K. are shareholders of curantis UG (Ltd). C.R. is a shareholder of lero bioscience UG (Ltd). All other authors have no conflicts of interest to disclose.

Funding Information

This work was supported by Deputy Vice-Chancellor Research (DVCR) start up grant to F.M.L. from the University of Sydney.

Supplementary Material

Supplementary Data

Supplementary Figure S1

Cite this article as: Couttas TA, Boost C, Pahlisch F, Sykorova EB, Leweke JE, Koethe D, Endepols H, Rohleder C, Leweke FM (2023) Simultaneous assessment of serum levels and pharmacologic effects of cannabinoids on endocannabinoids and N-acylethanolamines by liquid chromatography–tandem mass spectrometry, Cannabis and Cannabinoid Research 8:4, 657–669, DOI: 10.1089/can.2021.0181.

References

  • 1. Piomelli D. The molecular logic of endocannabinoid signalling. Nat Rev Neurosci. 2003;4:873–884. [DOI] [PubMed] [Google Scholar]
  • 2. Lu HC, Mackie K. An introduction to the endogenous cannabinoid system. Biol Psychiatry. 2016;79:516–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346:561–564. [DOI] [PubMed] [Google Scholar]
  • 4. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365:61–65. [DOI] [PubMed] [Google Scholar]
  • 5. Leweke FM, Giuffrida A, Wurster U, et al. Elevated endogenous cannabinoids in schizophrenia. Neuroreport. 1999;10:1665–1669. [DOI] [PubMed] [Google Scholar]
  • 6. Leweke FM, Mueller JK, Lange B, et al. Role of the endocannabinoid system in the pathophysiology of schizophrenia: implications for pharmacological intervention. CNS Drugs. 2018;32:605–619. [DOI] [PubMed] [Google Scholar]
  • 7. Schaefer C, Enning F, Mueller JK, et al. Fatty acid ethanolamide levels are altered in borderline personality and complex posttraumatic stress disorders. Eur Arch Psychiatry Clin Neurosci. 2014;264:459–463. [DOI] [PubMed] [Google Scholar]
  • 8. Jung KM, Astarita G, Yasar S, et al. An amyloid beta42-dependent deficit in anandamide mobilization is associated with cognitive dysfunction in Alzheimer's disease. Neurobiol Aging. 2012;33:1522–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Altamura C, Ventriglia M, Martini MG, et al. Elevation of plasma 2-arachidonoylglycerol levels in Alzheimer's disease patients as a potential protective mechanism against neurodegenerative decline. J Alzheimers Dis. 2015;46:497–506. [DOI] [PubMed] [Google Scholar]
  • 10. Pisani V, Moschella V, Bari M, et al. Dynamic changes of anandamide in the cerebrospinal fluid of Parkinson's disease patients. Mov Disord. 2010;25:920–924. [DOI] [PubMed] [Google Scholar]
  • 11. Rodriguez de Fonseca F, Navarro M, Gomez R, et al. An anorexic lipid mediator regulated by feeding. Nature. 2001;414:209–212. [DOI] [PubMed] [Google Scholar]
  • 12. Fidaleo M, Fanelli F, Ceru MP, et al. Neuroprotective properties of peroxisome proliferator-activated receptor alpha (PPARalpha) and its lipid ligands. Curr Med Chem. 2014;21:2803–2821. [DOI] [PubMed] [Google Scholar]
  • 13. Lambert DM, Vandevoorde S, Jonsson KO, et al. The palmitoylethanolamide family: a new class of anti-inflammatory agents? Curr Med Chem. 2002;9:663–674. [DOI] [PubMed] [Google Scholar]
  • 14. Pertwee RG, Howlett AC, Abood ME, et al. International union of basic and clinical pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB(1) and CB(2). Pharmacol Rev. 2010;62:588–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. O'Sullivan SE. An update on PPAR activation by cannabinoids. Br J Pharmacol. 2016;173:1899–1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Ahern GP. Activation of TRPV1 by the satiety factor oleoylethanolamide. J Biol Chem. 2003;278:30429–30434. [DOI] [PubMed] [Google Scholar]
  • 17. Ryberg E, Larsson N, Sjogren S, et al. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol. 2007;152:1092–1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Fonseca BM, Costa MA, Almada M, et al. Endogenous cannabinoids revisited: a biochemistry perspective. Prostaglandins Other Lipid Mediat. 2013;102–103:13–30. [DOI] [PubMed] [Google Scholar]
  • 19. Jonsson KO, Vandevoorde S, Lambert DM, et al. Effects of homologues and analogues of palmitoylethanolamide upon the inactivation of the endocannabinoid anandamide. Br J Pharmacol. 2001;133:1263–1275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Bisogno T, Maurelli S, Melck D, et al. Biosynthesis, uptake, and degradation of anandamide and palmitoylethanolamide in leukocytes. J Biol Chem. 1997;272:3315–3323. [DOI] [PubMed] [Google Scholar]
  • 21. Di Marzo V, Melck D, Orlando P, et al. Palmitoylethanolamide inhibits the expression of fatty acid amide hydrolase and enhances the anti-proliferative effect of anandamide in human breast cancer cells. Biochem J. 2001;358:249–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol. 2008;13:147–159. [DOI] [PubMed] [Google Scholar]
  • 23. Wollner HJ, Matchett JR, Levine J, et al. Isolation of a physiologically active tetrahydrocannabinol from cannabis sativa resin. J Am Chem Soc. 1942;64:26–29. [Google Scholar]
  • 24. Renard J, Krebs MO, Le Pen G, et al. Long-term consequences of adolescent cannabinoid exposure in adult psychopathology. Front Neurosci. 2014;8:361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Leweke FM, Giuffrida A, Koethe D, et al. Anandamide levels in cerebrospinal fluid of first-episode schizophrenic patients: impact of cannabis use. Schizophr Res. 2007;94:29–36. [DOI] [PubMed] [Google Scholar]
  • 26. Di Marzo V, Berrendero F, Bisogno T, et al. Enhancement of anandamide formation in the limbic forebrain and reduction of endocannabinoid contents in the striatum of delta9-tetrahydrocannabinol-tolerant rats. J Neurochem. 2000;74:1627–1635. [DOI] [PubMed] [Google Scholar]
  • 27. Rohleder C, Pahlisch F, Graf R, et al. Different pharmaceutical preparations of Delta(9)-tetrahydrocannabinol differentially affect its behavioral effects in rats. Addict Biol. 2020;25:e12745. [DOI] [PubMed] [Google Scholar]
  • 28. Leweke FM, Piomelli D, Pahlisch F, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2:e94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. McGuire P, Robson P, Cubala WJ, et al. Cannabidiol (CBD) as an adjunctive therapy in schizophrenia: a multicenter randomized controlled trial. Am J Psychiatry. 2018;175:225–231. [DOI] [PubMed] [Google Scholar]
  • 30. Rohleder C, Muller JK, Lange B, et al. Cannabidiol as a potential new type of an antipsychotic. A critical review of the evidence. Front Pharmacol. 2016;7:422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Elmes MW, Kaczocha M, Berger WT, et al. Fatty acid-binding proteins (FABPs) are intracellular carriers for Delta9-tetrahydrocannabinol (THC) and cannabidiol (CBD). J Biol Chem. 2015;290:8711–8721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Sparling PB, Giuffrida A, Piomelli D, et al. Exercise activates the endocannabinoid system. Neuroreport. 2003;14:2209–2211. [DOI] [PubMed] [Google Scholar]
  • 33. Katz PS, Sulzer JK, Impastato RA, et al. Endocannabinoid degradation inhibition improves neurobehavioral function, blood-brain barrier integrity, and neuroinflammation following mild traumatic brain injury. J Neurotrauma. 2015;32:297–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Piro JR, Suidan GL, Quan J, et al. Inhibition of 2-AG hydrolysis differentially regulates blood brain barrier permeability after injury. J Neuroinflammation. 2018;15:142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Navarrete F, Garcia-Gutierrez MS, Jurado-Barba R, et al. Endocannabinoid system components as potential biomarkers in psychiatry. Front Psychiatry. 2020;11:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Leweke FM, Mueller JK, Lange B, et al. Therapeutic potential of cannabinoids in psychosis. Biol Psychiatry. 2016;79:604–612. [DOI] [PubMed] [Google Scholar]
  • 37. Buczynski MW, Parsons LH. Quantification of brain endocannabinoid levels: methods, interpretations and pitfalls. Br J Pharmacol. 2010;160:423–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Keereetaweep J, Chapman KD. Lipidomic analysis of endocannabinoid signaling: targeted metabolite identification and quantification. Neural Plast. 2016;2016:2426398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Dong X, Li L, Ye Y, et al. Surrogate analyte-based quantification of main endocannabinoids in whole blood using liquid chromatography-tandem mass spectrometry. Biomed Chromatogr. 2019;33:e4439. [DOI] [PubMed] [Google Scholar]
  • 40. Balvers MG, Verhoeckx KC, Witkamp RF. Development and validation of a quantitative method for the determination of 12 endocannabinoids and related compounds in human plasma using liquid chromatography-tandem mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2009;877:1583–1590. [DOI] [PubMed] [Google Scholar]
  • 41. Jian W, Edom R, Weng N, et al. Validation and application of an LC-MS/MS method for quantitation of three fatty acid ethanolamides as biomarkers for fatty acid hydrolase inhibition in human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2010;878:1687–1699. [DOI] [PubMed] [Google Scholar]
  • 42. Lam PM, Marczylo TH, Konje JC. Simultaneous measurement of three N-acylethanolamides in human bio-matrices using ultra performance liquid chromatography-tandem mass spectrometry. Anal Bioanal Chem. 2010;398:2089–2097. [DOI] [PubMed] [Google Scholar]
  • 43. Schreiber D, Harlfinger S, Nolden BM, et al. Determination of anandamide and other fatty acyl ethanolamides in human serum by electrospray tandem mass spectrometry. Anal Biochem. 2007;361:162–168. [DOI] [PubMed] [Google Scholar]
  • 44. Liput DJ, Tsakalozou E, Hammell DC, et al. Quantification of anandamide, oleoylethanolamide and palmitoylethanolamide in rodent brain tissue using high performance liquid chromatography-electrospray mass spectroscopy. J Pharm Anal. 2014;4:234–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Giuffrida A, Rodriguez de Fonseca F, Piomelli D. Quantification of bioactive acylethanolamides in rat plasma by electrospray mass spectrometry. Anal Biochem. 2000;280:87–93. [DOI] [PubMed] [Google Scholar]
  • 46. Berman P, Futoran K, Lewitus GM, et al. A new ESI-LC/MS approach for comprehensive metabolic profiling of phytocannabinoids in Cannabis. Sci Rep. 2018;8:14280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Scheidweiler KB, Newmeyer MN, Barnes AJ, et al. Quantification of cannabinoids and their free and glucuronide metabolites in whole blood by disposable pipette extraction and liquid chromatography-tandem mass spectrometry. J Chromatogr. A 2016;1453:34–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Vozella V, Zibardi C, Ahmed F, et al. Fast and sensitive quantification of delta(9)-tetrahydrocannabinol and its main oxidative metabolites by liquid chromatography/tandem mass spectrometry. Cannabis Cannabinoid Res. 2019;4:110–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Kevin RC, Vogel R, Doohan P, et al. A validated method for the simultaneous quantification of cannabidiol, Delta(9) -tetrahydrocannabinol, and their metabolites in human plasma and application to plasma samples from an oral cannabidiol open-label trial. Drug Test Anal. 2021;13:614–627. [DOI] [PubMed] [Google Scholar]
  • 50. Zoerner AA, Gutzki FM, Batkai S, et al. Quantification of endocannabinoids in biological systems by chromatography and mass spectrometry: a comprehensive review from an analytical and biological perspective. Biochim Biophys Acta. 2011;1811:706–723. [DOI] [PubMed] [Google Scholar]
  • 51. U.S. Department of Health and Human Services, Food Drug Administration, Center for Drug Evaluation and Research (CDER), et al. Guidance for industry: bioanalytical method validation. 2001. [Google Scholar]
  • 52. Borman P, Elder D. Q2(R1) Validation of analytical procedures. In: ICH Quality Guidelines. 2017, pp. 127–166. [Google Scholar]
  • 53. Shah VP, Midha KK, Findlay JW, et al. Bioanalytical method validation—a revisit with a decade of progress. Pharm Res. 2000;17:1551–1557. [DOI] [PubMed] [Google Scholar]
  • 54. European Medicines Agency (EMEA). ICH Q2 (R1) Validation of analytical procedures: text and methodology—Step 5. Note for guidance on validation of analytical procedures: text and Methodologiy (CPMP/ICH/381/95). London, 1995. [Google Scholar]
  • 55. Hillard CJ, Weinlander KM, Stuhr KL. Contributions of endocannabinoid signaling to psychiatric disorders in humans: genetic and biochemical evidence. Neuroscience. 2012;204:207–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Hillard CJ. Circulating endocannabinoids: from whence do they come and where are they going? Neuropsychopharmacology. 2018;43:155–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Lanz C, Mattsson J, Stickel F, et al. Determination of the endocannabinoids anandamide and 2-arachidonoyl glycerol with gas chromatography-mass spectrometry: analytical and preanalytical challenges and pitfalls. Med Cannabis Cannabinoids. 2018;1:18–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Volkow ND, Fowler JS, Wang GJ, et al. Dopamine in drug abuse and addiction: results from imaging studies and treatment implications. Mol Psychiatry. 2004;9:557–569. [DOI] [PubMed] [Google Scholar]
  • 59. Gorelick DA. The rate hypothesis and agonist substitution approaches to cocaine abuse treatment. Adv Pharmacol (San Diego, Calif). 1998;42:995–997. [DOI] [PubMed] [Google Scholar]
  • 60. Oldendorf WH. Some relationships between addiction and drug delivery to the brain. NIDA Res Monogr. 1992;120:13–25. [PubMed] [Google Scholar]
  • 61. Zufferey F, Donze N, Rahban R, et al. Semen endocannabinoids are correlated to sperm quality in a cohort of 200 young Swiss men. Andrology. 2020;8:1126–1135. [DOI] [PubMed] [Google Scholar]
  • 62. Maccarrone M, Bari M, Lorenzon T, et al. Anandamide uptake by human endothelial cells and its regulation by nitric oxide. J Biol Chem. 2000;275:13484–13492. [DOI] [PubMed] [Google Scholar]
  • 63. Maccarrone M, Finazzi-Agro A. Endocannabinoids and their actions. Vitam Horm. 2002;65:225–255. [DOI] [PubMed] [Google Scholar]
  • 64. Scheidweiler KB, Schwope DM, Karschner EL, et al. In vitro stability of free and glucuronidated cannabinoids in blood and plasma following controlled smoked cannabis. Clin Chem. 2013;59:1108–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Rouzer CA, Ghebreselasie K, Marnett LJ. Chemical stability of 2-arachidonylglycerol under biological conditions. Chem Phys Lipids. 2002;119:69–82. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental data
Suppl_Data.docx (14.2KB, docx)
Supplemental data
Suppl_FigureS1.docx (262.8KB, docx)

Articles from Cannabis and Cannabinoid Research are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES