Abstract
Although methadone is effective in the management of acute pain, the complexity of its absorption-distribution-metabolism-excretion profile limits its use as an opioid of choice for perioperative analgesia. Because deuteration is known to improve the pharmacokinetic, pharmacodynamic and toxicological properties of some drugs, here we characterized the single dose pharmacokinetic properties and post-operative analgesic efficacy of d9-methadone.
The pharmacokinetic profiles of d9-methadone and methadone administered intravenously to CD-1 male mice revealed that deuteration lead to a 5.7- and 4.4-fold increase in the area under the time-concentration curve and maximum concentration in plasma, respectively, as well as reduction in clearance (0.9 ± 0.3 L/h/kg vs 4.7 ± 0.8 L/h/kg). The lower brain-to-plasma ratio of d9-methadone compared to that of methadone (0.35 ± 0.12 vs 2.05 ± 0.62) suggested that deuteration decreases the transfer of the drug across the blood-brain barrier. The estimated LD50 value for a single intravenous dose of d9-methadone was 2.1-fold higher than that for methadone. Moreover, d9-methadone outperformed methadone in the efficacy against postoperative pain by primarily activating peripheral opioid receptors. Collectively, these data suggest that the replacement of three hydrogen atoms in three methyl groups of methadone improved its pharmacokinetic properties, improved safety, and enhanced its analgesic efficacy.
Keywords: deuteration, d9-methadone, pharmacokinetic, metabolism, postoperative pain
1. Introduction
Methadone is a long-lasting synthetic opioid used as a maintenance therapy to treat opioid addiction and as a pain reliever. Recently, Kreutzwiser and Tawfic provided a comprehensive review article focusing on the use of methadone in the management of acute pain and chronic non-cancer and chronic cancer pain [1]. Accordingly, the existing data do not support the use of methadone as a first-line aid in non-cancer and cancer pain management [1]. On the other hand, several studies demonstrated the efficacy and safety of methadone in the management of acute pain during intra- and post-operative periods [2–6]. However, despite existing evidence that methadone decreases pain after surgery, the use of methadone as an opioid of choice for postoperative pain management is hindered by the complex absorption-distribution-metabolism-excretion profile of the drug [1].
The main challenges in the therapeutic use of methadone are associated with considerable inter-individual variation in its effect and toxicity. High inter-patient variability in the plasma concentration of methadone among those taking the same dose of the drug [7,8] is attributed mainly to variability in the rates of its hepatic and intestinal biotransformation [9–11]. Methadone undergoes sequential N-demethylation into its inactive derivatives 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine (EDDP) and 2-ethyl-5-methyl-3,3-diphenyl-1-pyrroline (EMDP) by several cytochrome P450 (CYP) enzymes, of which hepatic CYP3A4, CYP2B6, CYP2D6 and CYP2C19 are the major contributors [12]. The activity of these enzymes varies greatly among individuals due to genetic and environmental factors. While several reports showed correlation between functional genetic variants of methadone-metabolizing CYPs and the concentrations of methadone in plasma [12,13], it was later established that CYP2B6, a highly polymorphic CYP isoform, is the main determinant of clinical disposition of methadone in humans. Thus, a formulation of methadone that improves its pharmacokinetic (PK) and pharmacodynamic (PD) properties could potentially improve the clinical utility of this drug.
Deuterium is a naturally occurring, stable, non-radioactive hydrogen isotope. In recent years, the use of deuterated compounds in the clinical development of medications has become a fast-growing field on the premise of the proven safety of deuterium [14–16] and the ability of deuteration to improve the PK properties of drugs [17–25]. Previously, Elison et al. reported that the substitution of deuterium for hydrogen in the N-CH3 group of morphine resulted in a reduction in the rate of oxidative N-demethylation of d3-morphine [26]. On the other hand, Hsia et al. reported similar rates of absorption, distribution, and excretion of d3-methadone and methadone in rats, suggesting that the replacement of hydrogen atoms at C-1 with deuterium atoms in methadone did not affect its PK and PD properties [27]. These examples illustrate the differential effects of the incorporation of deuterium on the PK of drugs.
Therefore, the aims of this study were to evaluate whether the replacement of three hydrogen atoms in each of three methyl groups of methadone (Figure 1) affects the pharmacokinetic properties and organ distribution of d9-methadone following its single intravenous administration as well as its safety profile and analgesic potency.
Figure 1.

Chemical structures of d9-methadone and its metabolite d6-EDDP.
EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine.
2. Materials and Methods
2.1. Chemicals
Protein concentrations were measured using Bradford protein assay reagent (Bio-Rad Laboratories, Hercules, CA, USA) with bovine serum albumin as a standard. (±)-Methadone hydrochloride and (±)-d9-methadone (1 mg/ml solution in methanol) were purchased from Sigma-Aldrich (St. Louis, MO, USA). All other chemicals were purchased from Sigma-Aldrich (St. Louis, MO) unless otherwise mentioned.
2.2. Animals
All experimental procedures were approved by the Animal Care and Use Committee at the University of Texas Medical Branch (UTMB) at Galveston and were in accordance with guidelines published by the US National Institutes of Health Guide for the Care and Use of Laboratory Animals (NIH Publication No. 85–23, revised 1996). Hsd:ICR (CD-1) mice were purchased from Envigo (Indianapolis, IN) and C57BL/6N mice (7–10 weeks, both sexes) were purchased from Charles River Laboratories (Houston, TX). All animals were housed in temperature-controlled rooms (23°C) with a 12:12 hour light/dark cycle and with unlimited access to food and water in an American Association for Accreditation of Laboratory Animal Care (AAALAC)-accredited animal facility.
2.3. In vivo toxicity of d9-methadone vs methadone
The toxicity of d9-methadone and methadone administered intravenously (i.v.) via tail vein was assessed in CD-1 male mice using the Up-and-Down method [30]. Following the administration of the drug, each animal was observed for 20 minutes and scored using the Clinical Scores scale (1–5) as described in detail in our recent report [31]. We first evaluated the toxicity of methadone, for which the starting dose administered to the first animal was selected using a constant multiplicative factor of 1.2 and was five steps below 41 mg/kg; the latter value represented the LD50 of methadone which was administered intraperitoneally (i.p.) to Swiss mice [27]. The dose for the next animal was increased or decreased by a factor of 1.2 depending on whether the animal survived or died, respectively. The dosing was stopped when five reversals occurred in six animals tested consecutively. The estimated LD50 value was calculated using the maximum likelihood method imbedded in the AOT425 software v.1.0 (Westat, Inc. and U.S. Environmental Protection Agency, Washington, DC). The outcome was deemed the same for all animals with the Clinical Score of 5 for those sacrificed by CO2 asphyxiation for humane reasons and for those that had died during the testing. For d9-methadone, we utilized similar approach, however the starting dose was selected five steps above the LD50 value that we determined for methadone.
2.4. Pharmacokinetic and tissue distribution of d9-methadone vs methadone
The PK parameters of d9-methadone and methadone were determined following the i.v. administration in CD-1 male mice. N=3 experiments were conducted for each studied compound. For the PK study, (±)-methadone hydrochloride was diluted in Dulbecco’s phosphate buffered saline (DPBS, cell culture grade, Fischer Scientific, PA, USA) to achieve a final concentration of 0.5 mg/mL. To prepare (±)-d9-methadone solution, 5 mL of the 1 mg/ml stock in methanol, then the dried residue was reconstituted in 3 ml of 20 mM hydrochloric acid, and then PBS was added to achieve a final concentration of 0.5 mg/ml of d9-methadone. At the day of each experiment, each mouse (n=9 animals per experiment) received a 2 mg/kg dose of d9-methadone or methadone i.v. via tail vein. The dosage volume was adjusted based on a body weight of each mouse and was equivalent to 100 μL of the drug solution administered to a mouse weighing 25 g. At predetermined time points (5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 4 h, 6 h, and 8 h), n=1 animal was sacrificed by CO2 asphyxiation. The blood samples were obtained by cardiac puncture and centrifuged immediately to separate the plasma. The lungs, liver, spleen, heart, and a kidney were resected from each animal and weighed (for paired organs, both lungs were collected and processed together, while only one kidney was harvested from each animal). The plasma samples and the organs were stored at −80°C until analysis.
2.5. Tissue sample preparation
After thawing, the organs were weighed and homogenized with deionized water containing 1% formic acid. The ratios of the organ weight to the volume of deionized water (w/v) in the homogenates were: 1:1 for the brain, 1:3 for the liver, heart, lungs and kidney, and 1:4 for the spleen.
2.6. Sample Analysis
A liquid chromatography–mass spectrometry (LC-MS/MS) method for the quantification of methadone, d9-methadone, EDDP and d6-EDDP was developed and validated in our laboratory. The samples were analyzed with a Thermo Scientific™ TSQ Quantiva™ Triple Quadrupole mass spectrometer (Waltham, MA). The system was controlled by Xcalibur 3.0 software. Separation of the analytes was achieved by a Waters Symmetry C18 HPLC column at 45°C. The mobile phase was made of (A) acetonitrile and (B) 0.1% formic acid aqueous solution (v/v) with gradient elution of 0–5 min, 25% A to 50% A with a flow rate of 350 μL/min. Before each injection, the HPLC column was washed with 90% A, and then equilibrated with 25% A for 5 min. The following internal standards were utilized: d9-methadone (for quantitative determination of methadone), methadone (for d9-methadone), and d3-EDDP (for EDDP). Due to the unavailability of synthesized d6-EDDP as a standard compound, d3-EDDP was used in lieu of d6-EDDP to construct a calibration curve, with EDDP serving as internal standard. Multiple reaction monitoring (MRM) was set up at m/z 310→265 for methadone, m/z 319→268 for d9-methadone, m/z 278→249 for EDDP, m/z 284→249 for d6-EDDP, and m/z 281→249 for d3-EDDP.
The samples of plasma and tissue collected from the mice were processed as follows: 250 μL of plasma or 200 μL of organ homogenates were spiked with 10 μL of the corresponding internal standard working solution and 10 μL of Li2CO3 (10 mg/ml stock), and then the mixture was vortexed for 30 sec. Afterwards, 800 μL of acetonitrile was added and vortexed for 30 sec. The mixture was centrifuged at 12,000 × g for 15 min, and then the supernatant was transferred to a 2-mL centrifuge tube and dried at 50°C under a stream of nitrogen. The residue was reconstituted with 150 μL of initial mobile phase and centrifuged at 12,000 × g for 5 min. 100 μL of the supernatant was transferred to an HPLC injection vial and 10 μL of the sample was injected into the LC-MS/MS system.
The calibration and quality control standards of methadone, d9-methadone, EDDP and d3-EDDP (in lieu of d6-EDDP) were prepared by spiking the appropriate working solution of the standards with the blank plasma or the tissue homogenates, and the samples were processed as described above. The calibration curves for methadone, d9-methadone, EDDP and d3-EDDP were found to be linear in the ranges of 3.0–2400 ng/mL in plasma, 5.0–3900 ng/g in brain, 18.0–14000 ng/g in spleen, and 14.0–12000 ng/g in liver, kidney, lung and heart samples. The intra- and inter-day accuracy of the method was within 90% to 114%, and the precision was above 93%. All biological samples were processed and analyzed along with quality control samples at high, medium and low concentration levels.
2.7. Pharmacokinetic analysis
The PK parameters for methadone, d9-methadone, and their metabolites EDDP and d6-EDDP were determined using Kinetica software (v. 5.1, Thermo Fisher Scientific, Waltham, MA) [32–33]. The maximum plasma concentration (Cmax) and time to Cmax (Tmax) were determined by visual inspection of the concentration versus time curve for which the data for each experiment utilized multiple mice (one mouse per time point, total of n=3 experiments). The area under the plasma concentration vs. time curve over the 8-hour study period (AUC0–8h) was determined by non-compartmental analysis, utilizing the trapezoidal method. For methadone and d9-methadone, the clearance (CL) was calculated as the dose divided by the AUC0−∞, with the AUC0−∞ being calculated as the AUC0–8h plus an extrapolated AUC8h−∞ predicted as the concentration at 8 h divided by the terminal rate constant fit through the latter time points. The total volume of distribution (Vss) was calculated as the clearance multiplied by the mean residence time, which is the AUC0−∞ divided by the area under the moment curve (AUMC0−∞). The half-life was calculated as ln(2) divided by the terminal rate constant. The AUC0–8h for the parent drugs and their respective metabolites in each studied organ were derived using time vs. tissue concentration data from multiple mice (one animal per time point, total of n=3 experiments).
2.8. Metabolism of methadone and d9-methadone in vitro
The activity of mouse liver microsomes (MLM) and human liver microsomes (HLM) in catalyzing the N-demethylation of methadone to EDDP and d9-methadone to d6-EDDP was determined following the protocol described previously [28]. The stock solutions of methadone and d9-methadone were prepared as described above for the PK study, except that potassium phosphate buffer was used instead of PBS. Methadone and d9-methadone were used in a range of concentrations to construct saturation curves for the formation of their corresponding metabolites as follows: methadone, 7.5–100 μM with MLM and 15–300 μM with HLM; and d9-methadone, 1.875–60 μM with MLM and 7.5–100 μM with HLM. N=3 experiments were conducted for each drug, with each point in triplicate. Each reaction mixture (total volume of 0.2 mL in 100 mM potassium phosphate buffer, pH 7.4) containing 0.025 mg of pooled HLM (XenoTech, LLC, Kansas City, KS) or 0.05 mg of pooled MLM from male CD-1 mice (XenoTech, LLC, Kansas City, KS) and either methadone or d9-methadone was pre-incubated for 5 minutes at 37°C. The reaction was initiated by the addition of NADPH-regenerating system (0.4 mM NADP; 4 mM glucose-6-phosphate; 1U/mL glucose-6-phosphate dehydrogenase and 2 mM MgCl2) and carried out at 37°C. The duration of the reaction was 10 minutes for MLM and 20 minutes for HLM. The reaction was terminated by placement of the tubes on ice and the addition of 20 μL of Li2CO3 (10 mg/mL working solution) and 1.2 mL of ice-cold acetonitrile containing 1% formic acid to each tube as well as the respective internal standard (see section 2.6). The samples were processed as described in section 2.6; the final residues were reconstituted with 200 μL of the initial mobile phase and analyzed by means of LC-MS/MS for the quantitative determination of either EDDP or d6-EDDP as described in section 2.6. For each concentration of methadone and d9-methadone, the control reactions were conducted in the absence of NADPH-regenerating system, and the background quantities of EDDP and d6-EDDP were subtracted from the respective EDDP and d6-EDDP quantities determined in the reactions with NADPH. To minimize the number of freeze/thaw cycles of the liver microsomal proteins, the pooled microsomes were aliquoted and stored at −80°C as recommended by the manufacturer, and individual aliquots were defrosted for each experiment. The mouse and human liver microsomes utilized in this work were from the same respective lots to eliminate the influence of potential donor-associated variability in the activity of the hepatic enzymes. The experimental conditions were optimized using 30 μM of methadone for the MLM and HLM protein amounts and incubation time to assure that the formation of EDDP was within the linear range (assuming that the formation of d6-EDDP from d9-methadone was also linear under the same experimental conditions). The apparent Michaelis constant (Km) and maximum velocity (Vmax) values were estimated using SigmaPlot software, version 14.5.0.101 (Systat Software, Inc., Palo Alto, CA) using pooled data from n=3 saturation curves for each of the substrates. The intrinsic metabolic clearance CLint was calculated as Vmax/Km [29]; and the deuterium effect on the reaction was evaluated based on a ratio of Vmax and CLint parameters for the non-deuterated substrate to those of the deuterated substrate.
2.9. Animal models of pain
To model postoperative pain in mice, plantar incision was performed under isoflurane anesthesia. Specifically, a 4–8 mm incision was made through the skin, fascia, and muscle of the plantar side of one hind paw, longitudinally starting from the proximal edge of the heel. The skin was apposed with sutures (Fine Science Tools, Foster City, CA). The suture was removed at the end of postoperative day 2.
2.10. Behavioral testing
Mechanical pain sensitivity after plantar incision was assessed by measuring paw withdrawal behaviors in response to von Frey filament (VFF) probing. A mouse was placed in a plastic chamber on top of a mesh screen platform. The VFF of 0.98 mN was applied perpendicularly to the hind paw. At baseline (i.e., before surgery), mice rarely withdraw their hind paw from this VFF probing, whereas after surgery, the operated hind paw develops nociceptive hypersensitivity manifested as the increased number of withdrawals from ten trials of probing [34–35]. The percent of withdrawal responses out of the ten trials was recorded.
Methadone (3 mg/kg) or d9-methadone (3 mg/kg) was administered i.p. either once before plantar incision (i.e., intraoperative treatment) or repeatedly at 4-hour, 1-day, 2-day, and 3-day time points post-surgery (i.e., postoperative treatments). In a separate experiment that included the postoperative treatment, naloxone methiodide (NLXM, 10 mg/kg) was injected i.p. 30 minutes before the administration of either methadone or d9-methadone at 1-day post-surgery.
2.11. Statistical analysis
For the metabolism of methadone and d9-methadone by human liver microsomes in vitro, the Km and Vmax parameters were estimated for each substrate using pooled data from n=3 saturation curves (each data point represented the average of n=3 values from three saturation curves), and were compared descriptively. Likewise, CLint values were derived from the mean Km and Vmax values and compared descriptively.
The PK parameters for methadone and its metabolite EDDP were compared to the corresponding PK parameters of d9-methadone and its metabolite d6-EDDP by the Student’s t-test using Excel 2016 (Microsoft, Redmond, WA). P-values <0.05 were considered significant. The power of analysis was estimated post-hoc using G-power software, version 3.1 [36].
For sample size calculation in the behavioral study, we conducted preliminary experiments using three mice of plantar incision model in each of intra- and post-operative treatment regimens (both sexes at 1:2 ratio). The preliminary intra-operative treatment resulted in the Cohen’s d value of 2.1 between the methadone and d9-methadone groups in comparing the % of withdrawals measured 4 hours post-incision, indicating that at least five mice per group are needed to achieve statistical power of 0.8 at α=0.05 in this experimental setup (both sexes at 2:3 ratio). In the postoperative treatment experiment comparing % withdrawals before and after the treatment with d9-methadone (i.e., a paired comparison), Cohen’s d was >20, requiring at least two mice per group for the statistical power of 0.80 at α=0.05. Thus, we used n=3 mice per group for this experimental design (both sexes at 1:2 ratio). We used a generalized linear mixed model to analyze the number of withdrawals out of 10 trials (binomial distribution) repeatedly measured at multiple time points (AR1 covariance structure) with random intercepts for subject (i.e., mouse) variances. Degrees of freedom were allowed to vary across tests by Satterthwaite approximation.
3. Results
3.1. In vivo toxicity of d9-methadone vs methadone.
The estimated LD50 value for a single intravenous dose of methadone and d9-methadone were 11.6 mg/kg, 95% CI (11.5, 13.4) and 24.8 mg/kg, 95% CI (20.6, 28.7), respectively.
3.2. Pharmacokinetic and tissue distribution of d9-methadone vs. methadone.
Figures 2A–D show the plasma concentrations of methadone and d9-methadone and their corresponding metabolites EDDP and d6-EDDP as a function of time following i.v. administration of the studied drugs in male CD-1 mice. The plasma concentrations of both methadone and d9-methadone declined in a biexponential manner; the calculated PK parameters are presented in Table 1. The exposure to d9-methadone (AUC0–8h) and the Cmax of d9-methadone exceeded those of methadone by 6 folds (P < 0.05) and 4 folds (P < 0.0001), respectively. The Vss and CL values of d9-methadone were reduced by 8 and 5 folds (P < 0.01), respectively, as compared to methadone, while the difference in the elimination half-lives for both drugs did not attain statistical significance. EDDP and d6-EDDP, the corresponding metabolites of methadone and d9-methadone, were determined in the plasma of mice over the entire study period Figures 2C & 2D. The Cmax of d6-EDDP in plasma was 5-fold lower than the Cmax of EDDP (P < 0.001), while the difference between the Tmax values of the metabolites did not attain statistical significance (Table 1). The AUC0–8h of d6-EDDP was 2-fold lower than the AUC0–8h of EDDP (P < 0.01).
Figure 2.

Plasma concentration-time profiles for methadone and d9-methadone on a linear scale (A) and logarithmic scale (B) and their metabolites EDDP and d6-EDDP on a linear scale (C) and logarithmic scale (D) after intravenous administration to male CD-1 mice. Each mouse was administered 2 mg/kg of d9-methadone or methadone intravenously via the tail vein. At the predetermined time points of 5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 4 h, 6 h, and 8 h, animals were sacrificed by CO2 asphyxiation and blood samples were obtained by cardiac puncture. The quantification of methadone, d9-methadone, EDDP and d6-EDDP in plasma samples was performed using an LC-MS/MS method. Data are presented as the mean ± s.d. of 3 separate experiments using 1 animal per time point. N=3 animals were used for each time point. EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine.
Table 1.
Pharmacokinetic parameters of methadone and d9-methadone, and their respective metabolites, EDDP and d6-EDDP, in the plasma of male CD-1 mice after a single intravenous injection of the studied drugs.
| Parent Drug Parameters | Methadone | d9-Methadone | P-value |
|---|---|---|---|
| Dose (mg/kg) | 2.0 | 2.0 | — |
| Cmax (ng/mL) | 521 ± 55 | 2270 ± 181 | 0.0001 |
| Tmax (h) | 0.08 ± 0 | 0.08 ± 0 | — |
| AUC0–8h (ng·h/mL) | 414 ± 67 | 2362 ± 785 | 0.0128 |
| AUC0−∞ (ng·h/mL) | 436 ± 75 | 2389 ± 817 | 0.0146 |
| CL (L/h/kg) | 4.7 ± 0.8 | 0.9 ± 0.3 | 0.0019 |
| Vss (L/kg) | 8.9 ± 2.0 | 1.2 ± 0.0 | 0.0028 |
| Half-life (h) | 2.2 ± 0.8 | 1.1 ± 0.2 | 0.0796 |
| Metabolite Parameters | EDDP | d6-EDDP | P-value |
| Cmax (ng/mL) | 233 ± 28 | 48 ± 7 | 0.0004 |
| Tmax (h) | 0.14 ± 0.10 | 0.33 ± 0.14 | 0.1242 |
| AUC0–8h (ng·h/mL) | 257 ± 29 | 138 ± 25 | 0.0058 |
Methadone and d9-methadone, and their respective metabolites, EDDP and d6-EDDP, were quantified in the plasma samples of male CD-1 mice following a single intravenous injection of 2.0 mg/kg of the parent drug. The plasma concentration-time profiles of the drugs and their metabolites are shown in Figure 2. The PK parameters were determined using Kinetica software (v. 5.1). The data are presented as the mean ± s.d. of three experiments. EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine.
The ratio of the AUC0–8h of the metabolite to the AUC0–8h of the parent drug was lower for d9-methadone (0.06 ± 0.01) than for methadone (0.62 ± 0.04, P < 0.0001).
Following i.v. administration of the studied drugs, the extent of the exposure of the organs to d9-methadone ranked as following: lungs > kidneys ≈ liver > spleen > heart > brain, while the rank order for methadone was: kidneys ≈ lungs > spleen > liver > heart > brain. The exposure of the lungs and liver of the mice to d9-methadone was greater than the exposure to methadone, while the exposure of the heart, kidneys, and spleen to d9-methadone was lower than that of methadone (Table 2, P < 0.05). For all the results of the PK and tissue distribution experiments that were statistically significant by the standards of this study, the achieved statistical power was above 0.85 at α=0.05.
Table 2.
AUC0–8h of methadone, d9-methadone, and their respective metabolites EDDP and d6-EDDP in organs of male CD-1 mice after a single intravenous injection of 2.0 mg/kg of either methadone or d9-methadone.
| Parent Drug Tissue AUC0–8h (ng·h/g) | Methadone | d9-Methadone | P-value |
|---|---|---|---|
| Brain | 877 ± 395 | 780 ± 73 | 0.6971 |
| Liver | 3272 ± 570 | 5052 ± 117 | 0.0061 |
| Lung | 7694 ± 460 | 10799 ± 969 | 0.0074 |
| Heart | 1890 ± 115 | 1426 ± 230 | 0.0351 |
| Kidney | 7784 ± 469 | 5110 ± 408 | 0.0017 |
| Spleen | 4841 ± 494 | 3451 ± 148 | 0.0095 |
| Metabolite Tissue AUC0–8h (ng·h/g) | EDDP | d6-EDDP | P-value |
| Brain | 8.96 ± 5.26 | <LLOQ§ | <0.05 |
| Liver | 5608 ± 218 | 4375 ± 513 | 0.0186 |
| Lung | 9672 ± 856 | 9886 ± 614 | 0.7433 |
| Heart | 2155 ± 144 | 1243 ± 254 | 0.0057 |
| Kidney | 2815 ± 226 | 1562 ± 207 | 0.0021 |
| Spleen | 622 ± 45 | 411 ± 33 | 0.0028 |
The data reflect the exposure of individual organs to the parent drug and its metabolite and are presented as the mean ± s.d. of three experiments.
The concentration of d6-EDDP in the brain was below the lower limit of quantitation at every time point for all three animals. AUC, area under the tissue concentration vs. time curve; EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine; LLOQ, lower limit of quantification.
The ratios of AUC0–8h in the organs to the AUC0–8h in the plasma indicated substantial distribution and exposure of the mouse organs to methadone in comparison to reduced distribution ratios for d9-methadone (Figure 3, P < 0.01 for all organs). Despite similar exposure of the brain to d9-methadone and methadone (as reflected by similar AUC values), the brain-to-plasma AUC ratio of d9-methadone was 0.35 ± 0.12 while the brain-to-plasma AUC ratio for methadone was 2.05 ± 0.62 (Figure 3, P < 0.01), suggesting lower proportional transfer of d9-methadone across the blood-brain barrier (BBB).
Figure 3.

Ratio of AUC0–8h of methadone or d9-methadone in different organs to the AUC0–8h in plasma.
Each mouse was administered 2 mg/kg of d9-methadone or methadone intravenously via the tail vein. At predetermined time points (5 min, 15 min, 30 min, 1 h, 2 h, 3 h, 4 h, 6 h, and 8 h), n=1 animal per time point was sacrificed by CO2 asphyxiation, and the tissue samples were collected. The quantification of methadone and d9-methadone in tissue samples was performed using an LC-MS/MS method. AUC values for methadone and d9-methadone in each organ were determined using Kinetica software (v. 5.1). Data are presented as the mean + s.d. of 3 separate experiments using 1 animal per time point. N=3 animals were used for each time point.
AUC, area under the concentration vs. time curve. ** P < 0.01; *** P < 0.001.
The extent of the exposure of the organs to d6-EDDP and EDDP, the respective metabolites of d9-methadone and methadone, ranked as follows: lungs > liver > kidney ≈ heart > spleen. The exposure of the lungs to d6-EDDP and EDDP was similar, while the exposure to d6-EDDP in the liver, heart, kidney and spleen was significantly lower than the exposure to EDDP (Table 2, P<0.05). The concentration of d6-EDDP in the brain was below the lower limit of quantification (LLOQ).
3.3. Metabolism of methadone and d9-methadone.
The rates of the formation of EDDP and d6-EDDP from methadone and d9-methadone, respectively, were dependent on the substrate concentrations and exhibited typical saturation kinetics (Figure 4A–D). The apparent Km and Vmax values are shown in Table 3. The apparent Vmax of the formation of d6-EDDP from d9-methadone by MLM was 2.5-fold lower than the Vmax of the formation of EDDP from methadone. Meanwhile, the apparent Km of d9-methadone was 2.6-fold lower than the Km of methadone. Similar results were obtained using HLM: the estimated apparent Km and Vmax values for methadone N-demethylation were 2- and 2.5-folds lower than the corresponding values estimated for d9-methadone N-demethylation (Table 3). Thus, the deuteration effects of the drug’s N-demethylation Vmax (HVmax/DVmax) and CLint (HCLint/DCLint) were 2.5 and 1.3, respectively, in MLM, and 2.5 and 1, respectively, in HLM, and were deemed as isotope effects of low magnitude (<3.6) according to Miwa and Walsh [37].
Figure 4.

N-demethylation of methadone and d9-methadone into their respective metabolites, EDDP (A,C) and d6-EDDP (B,D) in vitro.
N-demethylation of methadone and d9-methadone into their respective metabolites, EDDP and d6-EDDP were tested in vitro using mouse liver microsomes (MLM) (A and B, respectively) and human liver microsomes (HLM) (C and D, respectively). The substrates were used in the following ranges: methadone, 7.5–100 μM with MLM and 15–300 μM with HLM; and d9-methadone, 1.875–60 μM with MLM and 7.5–100 μM with HLM. The substrates were incubates with the microsomal proteins at pH 7.4 in the presence of NADPH and in the absence of the cofactor (control reactions) at 37°C for either 10 min (MLM) or 20 min (HLM). The CYP-catalyzed formation of EDDP and d6-EDDP was calculated by subtracting the metabolite quantities determined in the control reactions from those determined in the reactions with NADPH. The rates of the formation of EDDP and d6-EDDP were dependent on the substrate concentrations and were fitted into the Michaelis-Menten model using SigmaPlot (version 14.5.0.101 for Windows, Systat Software Inc.). The insets show the corresponding Eadie-Hofstee plots. Data are presented as the mean ± s.d. from n=3 experiments, with each point in triplicate. The estimated kinetic parameters of the reactions are shown in Table 1. [V], reaction velocity, pmol/min/mg.protein; [S], substrate concentration, μM; EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine; MLM, mouse liver microsomes; HLM, human liver microsomes.
Table 3.
Kinetic parameters of the formation of EDDP and d6-EDDP from methadone and d9-methadone, respectively, by mouse and human liver microsomes.
| Subcellular fraction | Substrate | Km (μM) | Vmax (pmol·min−1·mg of protein−1) | CLint (Vmax/Km) | ||
|---|---|---|---|---|---|---|
| Best fit value | 95% CI | Best fit value | 95% CI | |||
| MLM | Methadone | 53.5 ± 5.1 | (42.3, 64.6) | 2631 ± 121 | (2368, 2893) | 49.2 |
| d 9 -Methadone | 20.7 ± 2.1 | (16.2, 25.2) | 1033 ± 45 | (937, 1129) | 49.7 | |
| HLM | Methadone | 82.9 ± 3.7 | (74.7, 91.0) | 1116 ± 17 | (1080, 1152) | 13.5 |
| d 9 -Methadone | 41.3 ± 5.9 | (28.4, 54.1) | 438 ± 27 | (379, 497) | 10.6 | |
The effect of the range of methadone and d9-methadone concentrations on the respective formation of EDDP and d6-EDDP was examined in mouse liver microsomes (MLM) and human liver microsomes (HLM) (Figure 4). The kinetic parameters were estimated by fitting the data in the Michaelis-Menten model using SigmaPlot software (version 14.5.0.101, Systat Software Inc., Palo Alto, CA). Data are presented as the mean ± S.E. of n=3 experiments.
EDDP, 2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine; HLM, human liver microsomes; MLM, mouse liver microsomes; Km, Michaelis constant; Vmax, maximum velocity; CI, confidence interval; CLint, intrinsic metabolic clearance.
3.4. Analgesic efficacy of d9-methadone against postoperative pain in mouse models.
Methadone has been shown to decrease postoperative pain with either intra- or post-operative treatment regimens [38–40]. As shown in Figure 5, mice treated with 3 mg/kg of d9-methadone demonstrated lower postoperative mechanical hypersensitivity on the day of surgery than methadone-treated mice (t(17)=3.37, P=0.004 at 2 hr; t(17)=2.33, P=0.033 at 4 hr). On the next day, however, there was no difference in the degree of postoperative mechanical hypersensitivity between the two groups (t(17)=0.33, P=0.75).
Figure 5.

The magnitude of postoperative mechanical pain hypersensitivity after intra-operative treatment with methadone or d9-methadone.
Using an intra-operative regimen, we tested if d9-methadone produces greater relief of postoperative pain than methadone. Under anesthesia, immediately before plantar incision, mice received either methadone or d9-methadone (3 mg/kg, i.p.) as an intra-operative treatment. Data are presented as the mean ± s.d. of 3 experiments. N=3 animals were used for each time point.
* P < 0.05; ** P < 0.01.
Methadone given postoperatively at a dose of 3 mg/kg was unable to reduce the postoperative mechanical hypersensitivity (Figure 6A). On the other hand, the same dose of d9-methadone alleviated hypersensitivity after each systemic injection once daily for 4 days (Figure 6B: F(3,22)=18.1, P<0.001 between times post-injection). Of note, no development of the apparent analgesic tolerance was observed in the ‘once daily for 4 days’ treatment regimen applied in this study (F(3,20)=0.335, P=0.8 between treatment days).
Figure 6.

The magnitude of post-operative mechanical pain hypersensitivity after post-operative treatment.
Using a post-operative regimen, we tested if d9-methadone produces greater relief of postoperative pain than methadone. After plantar incision, mice received 3 mg/kg, i.p., of either (A) methadone or (B) d9-methadone as a post-operative treatment. (C) Effect of naloxone methiodide (NLXM), a peripherally restricted opioid receptor antagonist, on the magnitude of post-operative mechanical pain hypersensitivity produced by d9-methadone. NLXM was administered postoperatively and 30 minutes prior to d9-methadone injection. NLXM completely blocked the effect of d9-methadone Data are presented as the mean ± s.d. of 3 experiments. N=3 animals were used for each time point.
As methadone’s analgesic effect is stringently mediated by opioid receptors in the periphery [41], we next investigated if the relief of postoperative pain by d9-methadone would be similarly mediated by peripheral opioid receptors. Naloxone methiodide (NLXM), a peripherally restricted opioid receptor antagonist, was administered to the operated animals 30 minutes before the treatment with d9-methadone. As a result, NLXM completely blocked the effect of d9-methadone (Figure 6C).
4. Discussion
It is estimated that postoperative pain occurs in 20 to 40% of surgical procedures, and pain following surgery remains one of the leading adverse outcomes that impacts postoperative recovery. Unfortunately, the use of repeated i.v. doses (i.e., i.v. boluses) of short-acting opioids such as morphine, oxycodone, and fentanyl result in fluctuating blood concentrations of opioid analgesics and, consequently, relatively brief periods of adequate pain relief. An alternative approach to the postoperative use of short-acting opioids is the use of a single-dose of a long-acting opioid such as methadone. However, the extensive first-pass metabolism of methadone as well as high inter-individual variability in its biotransformation (specifically, its N-demethylation) [42–45] diminish the suitability of methadone in the management of postoperative pain. Therefore, a formulation of methadone that decreases N-demethylation of methadone could potentially improve the clinical utility of this drug.
Deuterium modification has a potential to alter the pharmacokinetic parameters of a compound. A successfully designed deuterated compound may demonstrate an improvement over the parent (nondeuterated) compound by exhibiting increased AUC, increased Cmax and half-life, as well as reduced [46]. Therefore, the first aim of this study was to determine the effect of deuteration on the PK parameters and organ distribution of d9-methadone following its single i.v. administration to male CD-1 mice in comparison to nondeuterated methadone. The data obtained in this investigation revealed that the replacement of three hydrogen atoms in three methyl groups of methadone affected its PK properties. The AUC0–8h and Cmax of d9-methadone were greater than the AUC0–8h and Cmax of methadone, and the clearance of d9-methadone was lower than that of methadone. The decreased Vss for d9-methadone suggested that this deuterated analog of methadone remains mainly in the plasma, which is in agreement with its increased Cmax and AUC0–8h values in the plasma. Furthermore, the AUC0–8h value determined for the metabolite, d6-EDDP, demonstrated that deuteration of methadone also decreased the formation of d6-EDDP and consequently resulted in a decreased metabolite-to-parent drug ratio. The observed reduction in the clearance of d9-methadone can in part be explained by the reduced metabolic rate of the hepatic N-demethylation of the deuterated compound. Interestingly, the reduced clearance of d9-methadone was not accompanied by an increase in half-life. The reduced clearance accompanied by the reduced volume of distribution for d9-methadone may be explained by the differences in the extent of metabolism, differences in permeability across biological membranes, or by differences in binding to plasma proteins, which can all influence the extravascular/tissue distribution of the drug. An increase in plasma protein binding for d9-methadone compared to methadone would be expected to result in a decrease in the volume of distribution for d9-methadone. However, since changes in the unbound fraction in plasma do not always reflect the changes in the unbound fraction in tissue, a side-by-side evaluation of plasma protein binding of d9-methadone and methadone along with tissue binding would yield comparison of the unbound PK parameters and will be addressed in a future investigation.
The PK study revealed that the levels of d9-methadone and methadone in the brain were similar; however, the brain-to-plasma ratio for d9-methadone was 5.8-fold lower than the brain-to-plasma ratio of methadone. We determined that in mice the brain exposure to methadone exceeded the systemic exposure by 2-fold (brain-to-plasma ratio was 2.05), which was in agreement with previous studies reporting the brain-to-plasma ratio of 2.7 in rats [47] and brain-to-plasma ratios of 2.85 [48] and 2.3 [49] in humans determined postmortem. On the other hand, the AUC of d9-methadone in the brain of mice was lower than that in the plasma (brain-to-plasma ratio of 0.35), suggesting that deuteration decreased the permeability of methadone across the BBB. In principle, the polarity of deuterated compounds increases with the extent of deuterium-for-hydrogen substitution in the C-H bond [50]. Thus, decreased permeability of d9-methadone across biological membranes including the BBB would be expected. On the other hand, it is well known that P-glycoprotein (P-gp, ABCB1) limits the delivery of clinically used racemic methadone across the BBB [51]. Thus, it appears that the effect of deuteration on the lipophilicity and on the interaction of d9-methadone with P-gp are the likely reasons of the lower brain-to-plasma ratio of d9-methadone compared to that of methadone. To the best of our knowledge, there are no reports describing how incorporation of deuterium affects the permeability of drugs across biological membranes containing P-gp; thus, interaction of d9-methadone with P-gp should be further investigated.
Similar to the brain-to-plasma AUC ratio, the organ-to-plasma AUC ratios in the liver, lung, heart, kidney, and spleen for d9-methadone were also reduced several fold as compared to those of methadone (Figure 3), further suggesting the deuteration-associated changes in the proportion of the unbound and bound fractions of d9-methadone. In addition, the effect of deuteration on the interaction of d9-methadone with membrane efflux transporters of the organs could not be ruled out. The reduced exposure of the kidneys, spleen, and heart to d9-methadone as determined by the respective AUCs (Table 2) suggest decreased potential for d9-methadone to induce cardio- [52] and nephrotoxicity [53], which represent known adverse effects associated with the use of methadone. On the other hand, our study revealed that the exposure of the liver and lungs to d9-methadone was higher than to non-deuterated methadone. Since the treatment with methadone has not been linked to elevated serum enzymes or idiosyncratic acute liver injury [54], the administration of d9-methadone would not be expected to lead to hepatotoxicity.
As an opioid drug, methadone administered in excessive amounts can induce respiratory depression, which constitutes the major cause of death due to drug overdose in people taking prescription or illicit opioids [55]. Similar to humans, high doses of methadone can lead to respiratory depression in mice as well [55].
In this study, despite similar exposure of the murine brain to d9-methadone and methadone (Table 2), the estimated LD50 value for a single i.v. dose of d9-methadone was 2.1-fold higher than the estimated LD50 value for a single i.v. dose of methadone. Despite a multitude of pathophysiological studies of opioid-induced respiratory depression, Baldo et al. highlighted that the underlying mechanisms leading to the fatalities associated with opioid-induced respiratory depression are still not fully understood at this time [56]. Recent studies suggest that multiple sites of the brain can independently exert a depressive effect on breathing [56, 57], and the effect of d9-methadone on the μ opioid receptors of the inspiratory-generating regions of the brainstem should be investigated. Nevertheless, the comprehensive dose-dependent toxicokinetic assessment of d9-methadone is warranted. With respect to the main metabolite of d9-methadone, d6-EDDP, the exposure to d6-EDDP in the liver, heart, kidney and spleen was lower, while in the lungs it was similar to the pharmacologically inactive metabolite of methadone, EDDP.
The second aim of this study was to determine the effect of deuteration on the analgesic potency of d9-methadone in a model of postoperative pain. Because methadone’s analgesic effect is not mediated by central opioid receptors in the mouse [41] (in other words, the effect is strictly dependent on peripheral opioid receptors as is that of d9-methadone [Figure 6C]), it is expected that d9-methadone will be more potent in relieving pain than its nondeuterated counterpart by yielding a higher plasma concentration than methadone when administered at the same dose. The present study clearly supports this concept by showing that d9-methadone significantly relieved postoperative mechanical hypersensitivity in mice at a dose at which methadone was completely ineffective. Conversely, our results indicate that a greater dose of methadone would have been needed to achieve a similar degree of pain relief, elevating the concentration of the drug in the brain as well, which would consequently pose an increased risk for the central nervous system (CNS)-related side effects. And vice versa, lower doses of d9-methadone would be required to achieve a similar degree of pain relief caused by methadone. This could further lower the concentrations of deuterated drug in the brain and the organs included in the present study, thus decreasing the risk of possible adverse effects associated with the drug’s administration. Therefore, from the standpoint of developing effective and safe analgesics, our study demonstrated that the deuteration of methadone is a promising pharmacological approach for exploiting the proven involvement of the activated peripheral opioid receptors in the analgesic efficacy of systemically administered methadone. Meanwhile, the deuteration of methadone could reduce the undesired/negative consequences associated with the drug’s activation of opioid receptors in the brain.
Finally, we evaluated whether the substitution of three hydrogen atoms in three methyl groups of methadone would result in a reduced rate of the drug’s oxidative N-demethylation by mouse and human hepatic enzymes in vitro using MLM and HLM preparations. In principle, deuterium-for-hydrogen substitution in a C-H bond should increase the metabolic stability of a compound by decreasing the rate of its oxidative metabolism (C-D bonds are shorter and more stable to oxidative processes) [46]. Accordingly, our data demonstrated a 2.5-fold decrease in the rate of d9-methadone N-demethylation in vitro as compared to methadone in both MLM and HLM. The lower ratio of AUCmetabolite to AUCparent drug obtained for d9-methadone methadone in mice (0.06 ± 0.01 vs 0.62 ± 0.04, P < 0.0001) suggest the suitability of extrapolation to humans. The apparent Km of d9-methadone was 2- and 2.5-fold lower than that of methadone in humans and mice, respectively, suggesting higher affinity of d9-methadone to the hepatic CYP enzymes. The in vitro CLint of d9-methadone was 1.3-fold lower than the in vitro CLint of methadone, suggesting a rather modest effect of deuteration on the drug’s N-demethylation in vitro by human hepatic CYPs, while the N-demethylation of methadone by murine hepatic CYPs was not affected by the drug’s deuteration. It should be noted that the in vitro CLint values in the current study were obtained based on the formation of EDDP and might not reflect changes in the total CLint of the drug due to deuteration, presenting a limitation of the current study. While N-demethylation by hepatic CYP enzymes is the predominant mechanism of hepatic clearance of methadone in humans [58], the effect of deuteration on other metabolic pathways of the drug (and, consequently, the drug total CLint) could not be ruled out and will be addressed in a future investigation.
In summary, the data obtained in this investigation showed for the first time that deuterium replacement of three hydrogen atoms in three methyl groups altered the pharmacokinetic properties of methadone, improved its safety, and enhanced its analgesic effect in mice.
Acknowledgements
This work was supported by The Mildred Harvey and Phyllis Hankins Distinguished Chair in Obstetrics and Gynecology endowment (MSA) and Distinguished Research Chair in Obstetrics and Gynecology endowment (TN). The authors appreciate the financial support of Jerome Yaklic, Chair of the Department of Obstetrics and Gynecology at the University of Texas Medical Branch in Galveston. The analgesic efficacy experiments in this study (JL and JW) were supported by NIH R01 NS112344.
Abbreviations:
- ADME
absorption-distribution-metabolism-excretion
- AUC
area under the curve
- BBB
blood-brain barrier
- CL
clearance
- Vss
total volume of distribution
- Cmax
maximum plasma concentration
- CYP
cytochrome P450 enzymes
- EDDP
2-ethylidene-1,5-dimethyl-3,3,-diphenylpyrrolidine
- EMDP
2-ethyl-5-methyl-3,3-diphenyl-1-pyrroline
- HLM
human liver microsomes
- LC-MS/MS
liquid chromatography – mass spectrometry
- MLM
mouse liver microsomes
- MRM
Multiple reaction monitoring
- NLXM
naloxone methiodide
- PK
pharmacokinetic
- PD
pharmacodynamic
- Tmax
time of maximum plasma concentration
- VFF
von Frey filament
Footnotes
CRediT Authorship Contribution Statement
Xiao-ming Wang: Methodology, Validation, Formal analysis, Investigation, Writing - review & editing. Jigong Wang: Methodology, Validation, Formal analysis, Investigation, Writing - review & editing. Valentina Fokina: Validation, Formal analysis, Investigation, Writing – original draft. Svetlana Patrikeeva: Investigation, Data curation, Writing - review & editing. Erik Rytting: Formal analysis, Writing - review & editing. Mahmoud S. Ahmed: Conceptualization, Resources, Funding acquisition, Writing - review & editing. Jun-Ho La: Conceptualization, Methodology, Investigation, Writing – original draft, Supervision, Funding acquisition. Tatiana Nanovskaya: Conceptualization, Methodology, Investigation, Writing – original draft, Supervision, Funding acquisition.
Declaration of Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- [1].Kreutzwiser D, Tawfic QT. Methadone for Pain Management: A Pharmacotherapeutic Review. CNS Drugs 2020;34(8):827–839. doi: 10.1007/s40263-020-00743-3 [DOI] [PubMed] [Google Scholar]
- [2].Gottschalk A, Durieux ME, Nemergut EC. Intraoperative methadone improves postoperative pain control in patients undergoing complex spine surgery. Anesth Analg 2011;112(1):218–23. doi: 10.1213/ANE.0b013e3181d8a095 [DOI] [PubMed] [Google Scholar]
- [3].Murphy GS, Szokol JW, Avram MJ, Greenberg SB, Shear TD, Deshur MA, et al. Clinical Effectiveness and Safety of Intraoperative Methadone in Patients Undergoing Posterior Spinal Fusion Surgery: A Randomized, Double-blinded, Controlled Trial. Anesthesiology 2017;126(5):822–833. doi: 10.1097/ALN.0000000000001609 [DOI] [PubMed] [Google Scholar]
- [4].Murphy GS, Szokol JW, Avram MJ, Greenberg SB, Marymont JH, Shear T, et al. Intraoperative Methadone for the Prevention of Postoperative Pain: A Randomized, Double-blinded Clinical Trial in Cardiac Surgical Patients. Anesthesiology 2015;122(5):1112–22. doi: 10.1097/ALN.0000000000000633 [DOI] [PubMed] [Google Scholar]
- [5].Carvalho AC, Fábio Sebold FJG, Calegari PMG, de Oliveira BH, Trevisol FS. Comparison of postoperative analgesia with methadone versus morphine in cardiac surgery. Braz J Anesthesiol 2018;68(2):122–127. doi: 10.1016/j.bjan.2017.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Machado FC, Palmeira CCA, Torres JNL, Vieira JE, Ashmawi HA. Intraoperative use of methadone improves control of postoperative pain in morbidly obese patients: a randomized controlled study. J Pain Res 2018;11:2123–2129. doi: 10.2147/JPR.S172235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Holmstrand J, Anggard E, Gunne LM. Methadone maintenance: plasma levels and therapeutic outcome. Clin Pharmacol Ther 1978;23(2):175–80. doi: 10.1002/cpt1978232175 [DOI] [PubMed] [Google Scholar]
- [8].Tennant FS Jr. Inadequate plasma concentrations in some high-dose methadone maintenance patients. Am J Psychiatry 1987;144(10):1349–50. doi: 10.1176/ajp.144.10.1349 [DOI] [PubMed] [Google Scholar]
- [9].Moolchan ET, Umbricht A, Epstein D. Therapeutic drug monitoring in methadone maintenance: choosing a matrix. J Addict Dis 2001;20(2):55–73. doi: 10.1300/J069v20n02_05 [DOI] [PubMed] [Google Scholar]
- [10].Kharasch ED, Hoffer C, Whittington D, Sheffels P. Role of hepatic and intestinal cytochrome P450 3A and 2B6 in the metabolism, disposition, and miotic effects of methadone. Clin Pharmacol Ther 2004;76(3):250–69. doi: 10.1016/j.clpt.2004.05.003 [DOI] [PubMed] [Google Scholar]
- [11].Oda Y, Kharasch ED. Metabolism of methadone and levo-alpha-acetylmethadol (LAAM) by human intestinal cytochrome P450 3A4 (CYP3A4): potential contribution of intestinal metabolism to presystemic clearance and bioactivation. J Pharmacol Exp Ther 2001;298(3):1021–32. [PubMed] [Google Scholar]
- [12].Fonseca F, de la Torre R, Díaz L, Pastor A, Cuyàs E, Pizarro N, et al. Contribution of cytochrome P450 and ABCB1 genetic variability on methadone pharmacokinetics, dose requirements, and response. PLoS One 2011;6(5):e19527. doi: 10.1371/journal.pone.0019527 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Eap CB, Broly F, Mino A, Hämmig R, Déglon JJ, Uehlinger C, Meili D, et al. Cytochrome P450 2D6 genotype and methadone steady-state concentrations. J Clin Psychopharmacol 2001;21(2):229–34. doi: 10.1097/00004714-200104000-00016. [DOI] [PubMed] [Google Scholar]
- [14].Schoeller DA. Measurement of energy expenditure in free-living humans by using doubly labeled water. J Nutr. Nov 1988;118(11):1278–89. doi: 10.1093/jn/118.11.1278 [DOI] [PubMed] [Google Scholar]
- [15].Dufner D, Previs SF. Measuring in vivo metabolism using heavy water. Curr Opin Clin Nutr Metab Care 2003;6(5):511–7. doi: 10.1097/00075197-200309000-00004 [DOI] [PubMed] [Google Scholar]
- [16].Gant TG. Using deuterium in drug discovery: leaving the label in the drug. J Med Chem 2014;57(9):3595–611. doi: 10.1021/jm4007998 [DOI] [PubMed] [Google Scholar]
- [17].Schneider F, Hillgenberg M, Koytchev R, Alken RG. Enhanced plasma concentration by selective deuteration of rofecoxib in rats. Arzneimittelforschung 2006;56(4):295–300. doi: 10.1055/s-0031-1296724 [DOI] [PubMed] [Google Scholar]
- [18].Manley PW, Blasco F, Mestan J, Aichholz R. The kinetic deuterium isotope effect as applied to metabolic deactivation of imatinib to the des-methyl metabolite, CGP74588. Bioorg Med Chem 2013;21(11):3231–9. doi: 10.1016/j.bmc.2013.03.038 [DOI] [PubMed] [Google Scholar]
- [19].Xu G, Lv B, Roberge JY, et al. Design, synthesis, and biological evaluation of deuterated C-aryl glycoside as a potent and long-acting renal sodium-dependent glucose cotransporter 2 inhibitor for the treatment of type 2 diabetes. J Med Chem 2014;57(4):1236–51. doi: 10.1021/jm401780b [DOI] [PubMed] [Google Scholar]
- [20].Guo S, Pang X, Peng L, Zhan M, Fan L, Gong Y, et al. Design, synthesis and biological evaluation of deuterated Tivozanib for improving pharmacokinetic properties. Bioorg Med Chem Lett 2015;25(11):2425–8. doi: 10.1016/j.bmcl.2015.03.088 [DOI] [PubMed] [Google Scholar]
- [21].Pang X, Peng L, Chen Y. Effect of N-methyl deuteration on pharmacokinetics and pharmacodynamics of enzalutamide. J Labelled Comp Radiopharm 2017;60(9):401–409. doi: 10.1002/jlcr.3516 [DOI] [PubMed] [Google Scholar]
- [22].Velthuisen EJ, Baughman TM, Johns BA, Temelkoff DP, Weatherhead JG. Synthesis and pharmacokinetic profile of highly deuterated brecanavir analogs. Eur J Med Chem 2013;63:202–12. doi: 10.1016/j.ejmech.2013.02.001 [DOI] [PubMed] [Google Scholar]
- [23].Jiang J, Pang X, Li L, et al. Effect of N-methyl deuteration on metabolism and pharmacokinetics of enzalutamide. Drug Des Devel Ther 2016;10:2181–91. doi: 10.2147/DDDT.S111352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Stringer RA, Williams G, Picard F, Sohal B, Kretz O, McKenna J, et al. Application of a deuterium replacement strategy to modulate the pharmacokinetics of 7-(3,5-dimethyl-1H-1,2,4-triazol-1-yl)-3-(4-methoxy-2-methylphenyl)-2,6-dimethylp yrazolo[5,1-b]oxazole, a novel CRF1 antagonist. Drug Metab Dispos 2014;42(5):954–62. doi: 10.1124/dmd.114.057265 [DOI] [PubMed] [Google Scholar]
- [25].Harbeson SL, Morgan AJ, Liu JF, Aslanian A, Nguyen S, Bridson G, et al. Altering Metabolic Profiles of Drugs by Precision Deuteration 2: Discovery of a Deuterated Analog of Ivacaftor with Differentiated Pharmacokinetics for Clinical Development. J Pharmacol Exp Ther 2017;362(2):359–367. doi: 10.1124/jpet.117.241497 [DOI] [PubMed] [Google Scholar]
- [26].Elison C, Rapoport H, Laursen R, Elliott HW. Effect of deuteration of N--CH3 group on potency and enzymatic N-demethylation of morphine. Science 1961;134(3485):1078–9. doi: 10.1126/science.134.3485.1078 [DOI] [PubMed] [Google Scholar]
- [27].Hsia J, Tam J, Giles H, Leung CC, Marcus H, Marshman JA, et al. Markers for detection of supplementation in narcotic programs--deuterium-labeled methadone. Science 1976;193(4252):498–500. doi: 10.1126/science.941022 [DOI] [PubMed] [Google Scholar]
- [28].Fokina VM, Patrikeeva SL, Zharikova OL, Nanovskaya TN, Hankins GV, Ahmed MS. Transplacental transfer and metabolism of buprenorphine in preterm human placenta. Am J Perinatol 2011;28(1):25–32. doi: 10.1055/s-0030-1262508 [DOI] [PubMed] [Google Scholar]
- [29].Sharma R, Strelevitz TJ, Gao H, Clark AJ, Schildknegt K, Obach RS, et al. Deuterium isotope effects on drug pharmacokinetics. I. System-dependent effects of specific deuteration with aldehyde oxidase cleared drugs. Drug Metab Dispos 2012;40(3):625–34. doi: 10.1124/dmd.111.042770. [DOI] [PubMed] [Google Scholar]
- [30].OECD/OCDE. OECD Guideline for Testing of Chemicals: Acute Oral Toxicity- Up-and-Down Procedure. https://ntp.niehs.nih.gov/iccvam/suppdocs/feddocs/oecd/oecd_gl425-508.pdf
- [31].Fokina V, Patrikeeva S, Wang X, Shah M, Shah P, Russell W, Ahmed MS, Rytting E, Nanovskaya TN. Physicochemical and biological properties of membrane vesicles derived from human term placentas. Journal of Biomedical Nanotechnology 2022;18(2):589–599. doi: 10.1166/jbn.2022.3255. [DOI] [PubMed] [Google Scholar]
- [32].Rytting E, Nanovskaya TN, Wang X, Vernikovskaya DI, Clark SM, Cochran M, et al. Pharmacokinetics of indomethacin in pregnancy. Clinical pharmacokinetics 2014;53(6):545–51. doi: 10.1007/s40262-014-0133-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Rytting E, Wang X, Vernikovskaya DI, Zhan Y, Bauer C, Abdel-Rahman SM, et al. Metabolism and disposition of bupropion in pregnant baboons (Papio cynocephalus). Drug Metab Dispos 2014;42(10):1773–9. doi: 10.1124/dmd.114.058255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Hankerd K, McDonough KE, Wang J, Tang SJ, Chung JM, La JH. Postinjury stimulation triggers a transition to nociplastic pain in mice. Pain 2022;163(3):461–473. doi: 10.1097/j.pain.0000000000002366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].La JH, Wang J, Bittar A, Shim HS, Bae C, Chung JM. Differential involvement of reactive oxygen species in a mouse model of capsaicin-induced secondary mechanical hyperalgesia and allodynia. Mol Pain 2017;13:1744806917713907. doi: 10.1177/1744806917713907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Faul F, Erdfelder E, Buchner A, Lang AG. Statistical power analyses using G*Power 3.1: Tests for correlation and regression analyses. Behav. Res. Methods, 2009, 41, 1149–1160. doi: 10.3758/BRM.41.4.1149. doi: 10.3758/BRM.41.4.1149 [DOI] [PubMed] [Google Scholar]
- [37].Miwa GT, Walsh JS, Kedderis GL, Hollenberg PF. The use of intramolecular isotope effects to distinguish between deprotonation and hydrogen atom abstraction mechanisms in cytochrome P-450- and peroxidase-catalyzed N-demethylation reactions. J Biol Chem 1983;258(23):14445–9. [PubMed] [Google Scholar]
- [38].Kharasch ED. Intraoperative methadone: rediscovery, reappraisal, and reinvigoration? Anesth Analg 2011;112(1):13–6. doi: 10.1213/ANE.0b013e3181fec9a3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Gottschalk A, Durieux ME, Nemergut EC. Intraoperative methadone improves postoperative pain control in patients undergoing complex spine surgery. Anesth Analg 2011;112(1):218–23. doi: 10.1213/ANE.0b013e3181d8a095. [DOI] [PubMed] [Google Scholar]
- [40].Neto JO, Machado MD, de Almeida Correa M, Scomparim HA, Posso IP, Ashmawi HA. Methadone patient-controlled analgesia for postoperative pain: a randomized, controlled, double-blind study. J Anesth 2014;28(4):505–10. doi: 10.1007/s00540-013-1785-3. [DOI] [PubMed] [Google Scholar]
- [41].He L, Kim J, Ou C, McFadden W, van Rijn RM, Whistler JL. Methadone antinociception is dependent on peripheral opioid receptors. J Pain 2009;10(4):369–79. doi: 10.1016/j.jpain.2008.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Gadel S, Crafford A, Regina K, Kharasch ED. Methadone N-demethylation by the common CYP2B6 allelic variant CYP2B6.6. Drug Metab Dispos. 2013. Apr;41(4):709–13. doi: 10.1124/dmd.112.050625. Epub 2013 Jan 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Gadel S, Friedel C, Kharasch ED. Differences in Methadone Metabolism by CYP2B6 Variants. Drug Metab Dispos. 2015. Jul;43(7):994–1001. doi: 10.1124/dmd.115.064352. Epub 2015 Apr 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Crettol S, Déglon JJ, Besson J, Croquette-Krokkar M, Gothuey I, Hämmig R, Monnat M, Hüttemann H, Baumann P, Eap CB. Methadone enantiomer plasma levels, CYP2B6, CYP2C19, and CYP2C9 genotypes, and response to treatment. Clin Pharmacol Ther. 2005. Dec;78(6):593–604. doi: 10.1016/j.clpt.2005.08.011. [DOI] [PubMed] [Google Scholar]
- [45].Kharasch ED, Regina KJ, Blood J, Friedel C. Methadone Pharmacogenetics: CYP2B6 Polymorphisms Determine Plasma Concentrations, Clearance, and Metabolism. Anesthesiology. 2015. Nov;123(5):1142–53. doi: 10.1097/ALN.0000000000000867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Pirali T, Serafini M, Cargnin S, Genazzani AA. Applications of Deuterium in Medicinal Chemistry. J Med Chem 2019;62(11):5276–5297. doi: 10.1021/acs.jmedchem.8b01808 [DOI] [PubMed] [Google Scholar]
- [47].Liu SJ, Roerig DL, Wang RI. Brain and plasma levels of methadone and their relationships to analgesic activity of methadone in rats. Drug Metab Dispos 1983;11(4):335–8. [PubMed] [Google Scholar]
- [48].Iwersen-Bergmann S, Plattner S, Hischke S, et al. Brain/blood ratios of methadone and ABCB1 polymorphisms in methadone-related deaths. Int J Legal Med 2021;135(2):473–482. doi: 10.1007/s00414-021-02502-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Holm KM, Linnet K. Distribution of enantiomers of methadone and its main metabolite EDDP in human tissues and blood of postmortem cases. J Forensic Sci 2015;60(1):95–101. doi: 10.1111/1556-4029.12627. [DOI] [PubMed] [Google Scholar]
- [50].Wade D Deuterium isotope effects on noncovalent interactions between molecules. Chem Biol Interact. 1999. Feb 12;117(3):191–217. doi: 10.1016/s0009-2797(98)00097-0. [DOI] [PubMed] [Google Scholar]
- [51].Wang JS, Ruan Y, Taylor RM, Donovan JL, Markowitz JS, DeVane CL. Brain penetration of methadone (R)- and (S)-enantiomers is greatly increased by P-glycoprotein deficiency in the blood-brain barrier of Abcb1a gene knockout mice. Psychopharmacology (Berl) 2004;173(1–2):132–8. doi: 10.1007/s00213-003-1718-1 [DOI] [PubMed] [Google Scholar]
- [52].Alinejad S, Kazemi T, Zamani N, Hoffman RS, Mehrpour O. A systematic review of the cardiotoxicity of methadone. EXCLI J. 2015. May 5;14:577–600. doi: 10.17179/excli2015-553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Alinejad S, Ghaemi K, Abdollahi M, Mehrpour O. Nephrotoxicity of methadone: a systematic review. Springerplus. 2016. Dec 9;5(1):2087. doi: 10.1186/s40064-016-3757-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54]. https://www.ncbi.nlm.nih.gov/books/NBK548084/
- [55].Lewanowitsch T, Miller JH, Irvine RJ. Reversal of morphine, methadone and heroin induced effects in mice by naloxone methiodide. Life Sci. 2006. Jan 11;78(7):682–8. doi: 10.1016/j.lfs.2005.05.062. Epub 2005 Aug 15. [DOI] [PubMed] [Google Scholar]
- [56].Baldo BA. Current research in pathophysiology of opioid-induced respiratory depression, neonatal opioid withdrawal syndrome, and neonatal antidepressant exposure syndrome. Curr Res Toxicol. 2022. Jun 6;3:100078. doi: 10.1016/j.crtox.2022.100078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Bachmutsky I, Wei XP, Kish E, Yackle K. Opioids depress breathing through two small brainstem sites. Elife. 2020. Feb 19;9:e52694. doi: 10.7554/eLife.52694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Kharasch ED, Greenblatt DJ. Methadone Disposition: Implementing Lessons Learned. J Clin Pharmacol. 2019. Aug;59(8):1044–1048. doi: 10.1002/jcph.1427. Epub 2019 May 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
