Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2025 May 10;39(3):e70013. doi: 10.1111/fcp.70013

ABCB1, SLC22A1, COMT, and OPRM1 genotypes: Study of their influence on plasma methadone levels and clinical response to methadone maintenance treatment in opioid use disorder

Abd El Kader Ait Tayeb 1,2,, Edouard‐Jules Laforgue 3,4, Benoit Schreck 3,5, Marie Grall‐Bronnec 3,5, Jean‐Benoit Hardouin 3, Juliette Leboucher 5; OPAL Group
PMCID: PMC12065018  PMID: 40346879

Abstract

Background

Opioid use disorder (OUD) is an emerging and global public health concern, and its management remains inadequate, notably due to a lack of biomarkers, except for the CYP2B6 genetic polymorphisms.

Objectives

Hence, the aim of this study was to assess the influence of genetic polymorphisms of ABCB1, SLC22A1, COMT, and OPRM1 on biological parameters and clinical response in patients receiving methadone maintenance treatment (MMT).

Methods

A subgroup of 72 patients treated by MMT was genotyped for ABCB1 (rs1045642; rs2032582), SLC22A1 (rs12208357; rs72552763; rs113569197), COMT (rs4680), and OPRM1 (rs1799971) from Opioid PhArmacoLogy (OPAL), a clinical survey of patients suffering from OUD. Associations of these polymorphisms and both clinical and pharmacological (plasma methadone levels) responses were investigated.

Results

All polymorphisms tested were not associated with (R,S)‐methadone concentrations/doses (concentrations relative to doses), (R)‐methadone concentrations/doses nor (S)‐methadone concentrations/doses in bivariate analyses with codominant and recessive models. Also, polymorphisms tested were not related to clinical response (opiate cessation) during MMT in treated patients. The main limitations of our study were the sample size and the absence of polygenic analyses.

Conclusion

This study found no evidence to support the use of genotyping for polymorphisms in the ABCB1, SLC22A1, COMT, and OPRM1 genes in a clinical setting for the management of MMT in OUD.

Keywords: methadone maintenance treatment, opioid use disorder, pharmacodynamic targets, pharmacogenetics, pharmacokinetic targets


Abbreviations

CCTIRS

Advisory Committee on the Processing of Health Research Information

CNIL

Data Protection Commission

COMT

catechol‐O‐methyl‐transferase

CYP

cytochrome P450

EDDP

2‐ethylidene‐1,5‐dimethyl‐3,3‐diphenylpyrrolidine

FDA

US Food and Drug Administration

GNEDS

Local Research Ethics Committee

hERG

human voltage‐gated potassium channel

LC/MS‐MS

liquid chromatography coupled with mass spectrometry

MAF

minor allelic frequency

MMT

methadone maintenance treatment

OCT1

organic cation transporter 1

OPAL

Opioid Pharmacology

OUD

Opioid use disorder

1. INTRODUCTION

Opioid use disorder (OUD), along with the global opioid crisis, is a current and growing major public health concern with a prevalence of 510 people per 100 000 population (affecting around 40 million people worldwide) [1]. OUD could be defined as a pattern of opioid use (including heroin use, extra‐medical use of prescribed opioids, or use of illicitly manufactured synthetic opioids) associated with a range of mental, physical, social, and legal issues leading to clinically significant impairment [1, 2]. Its management, which has become a priority of public health policies, relies in part on the use of opioid maintenance treatment medication such as buprenorphine (a partial μ‐opioid receptor agonist and κ‐opioid receptor antagonist) or methadone (a full μ‐opioid receptor agonist) and on social and psychological interventions [1, 2, 3, 4].

Methadone, a ligand of the μ‐opioid receptor (a G‐protein coupled receptor coded by OPRM1), is a listed narcotic approved in France as a maintenance treatment for OUD [4, 5]. The methadone used in clinical practice is a racemic mixture of (R)‐ and (S)‐methadone. The (R)‐enantiomer seems to be associated with most of the opioid effect [6] and has a 10‐fold more potent agonist action on the μ‐opioid receptor [7], whereas (S)‐methadone is 3.5 times more potent than (R) in blocking the human voltage‐gated potassium channel (hERG) [8]. Hence, elevated (R)‐methadone may induce respiratory depression, whereas (S)‐methadone more potently blocks the hERG (related to long QT syndrome) [7, 8, 9].

The interindividual variability of methadone is important, and physicians have to titrate each patient progressively according to the clinical response [10]. Indeed, large interindividual variation exists in the response, tolerance to treatment, and onset of withdrawal symptoms, due to interindividual variability in pharmacokinetic (methadone blood level and elimination half‐life for a given dose) and pharmacodynamic parameters [6]. This variability could be related to pharmacogenetic biomarkers.

Methadone is intensively metabolized by the liver and intestine by several cytochromes P450 (CYP). Thus, our team and others previously showed the impacts of genetic polymorphisms of CYP2B6 on the methadone concentration [5, 6, 8, 11, 12, 13]. Other polymorphisms of drug‐metabolizing enzymes such as CYP2D6 [5, 14], and CYP3A4 [14, 15], and of carriers such as ABCB1 (P‐gp transporter) [11, 12, 16] have been highlighted to be associated with plasma methadone levels. The influence of genetic polymorphisms on the treatment response (illicit opiate cessation) has been less explored and is not yet supported by much evidence [6, 14, 17]. Interestingly, other carriers have been explored in prescribed opioids, such as the organic cation transporter 1 (OCT1), coded by SLC22A1, with the response to tramadol. OCT1 is primarily present on the sinusoidal membrane of hepatocytes and in the blood–brain barrier. It is responsible for the uptake of positively charged compounds at physiological pH, such as morphine, and the active metabolite of tramadol, O‐desmethyltramadol [18].

Concerning genes involved in pharmacodynamic targets or properties, associations between genetic polymorphisms and the response to methadone have been reported for OPRM1 [19, 20] and its biological cascade (such as ARRB2 coding for β‐arrestin 2) [21]. Obviously, associations were evidenced with polymorphisms of monoaminergic systems (dopamine D2 receptor (DRD2) [22], catechol‐O‐methyl‐transferase (COMT) [23] and functional polymorphisms related to synaptic levels of dopamine or serotonin [20]).

Somogyi et al concluded that the pharmacogenomics of methadone can be considered to reside in its various targets, especially the OPRM1 gene and its efflux transporter (ABCB1) [24]. This conclusion was further confirmed in a genome‐wide pharmacogenomics study [25]. These genetic elements contribute to the response or dosage stratification [26]. Nonetheless, publications summarizing previously studied pharmacological targets have concluded that drawing clear conclusions about the influence of these genes on the treatment response is currently impossible and that results should be interpreted with caution [27, 28].

In view of these misted results, our study aimed to evaluate the impacts of the ABCB1, SLC22A1, COMT, and OPRM1 polymorphisms on methadone enantiomer levels and clinical outcome to methadone maintenance treatment (MMT) in patients with OUD.

2. MATERIAL AND METHODS

2.1. Study oversight and patients

OPAL (Clinical trials: NCT01847729) was an observational transversal multicenter study involving 10 care centers in France which was approved by the local Research Ethics Committee (GNEDS), Advisory Committee on the Processing of Health Research Information (CCTIRS) and Data Protection Commission (CNIL). The sample consisted of 263 patients aged 18 years or older receiving maintenance treatment for OUD for at least 6 months. The primary objective was to determine the prevalence of addictive comorbidities [29]. The secondary objectives of the study included association studies aimed at identifying genetic biomarkers of methadone response in the context of OUD. Hence, the present work is an ancillary study performed solely on patients treated with methadone [5]. Thus, a total of 72 patients were included in this nested study.

2.2. Study procedure

The complete study procedure has been described and published previously [29]. Briefly, the clinical assessment included a clinical structured interview carried out by a physician during a medical consultation with the patient and a set of standardized self‐report questionnaires. Blood samples were collected immediately before administration from subjects who had been on MMT for at least 6 months without any change in dosage in the 5 days prior, to performing the ancillary study. Sociodemographic data included opioid dependence, data relating to the MMT, and psychopathological data. The characteristics of MMT were collected as follows: duration; MMT initiation with daily supervised dosing by a qualified health professional; maximal daily dose; current daily dose; compliance; withdrawal symptoms; current opioid use despite MMT or opioid abstinence, defined as the self‐reported absence of opioid use over the previous 6 months. The 6‐month period was chosen because it corresponds to the time usually required to stabilize the treatment dosage and because each patient could be asked about this period.

2.3. Variant selection

For each gene, variants were selected for this pharmacogenetic study using the PharmGKB database [30]. The selected variants had to be polymorphisms (MAF > 0.01), known to be associated with response to other treatments, and to impact the biological function of the protein or transporter. Thus, the chosen variants are: rs1045642 and rs2032582 for ABCB1; rs12208357, rs72552763 and rs113569197 for SLC22A1; rs4680 for COMT; rs1799971 for OPRM1.

2.4. Genotyping

The patients were genotyped for different genetic polymorphisms of ABCB1 (rs1045642; rs2032582), SLC22A1 (rs12208357; rs72552763; rs113569197), COMT (rs4680) and OPRM1 (rs1799971) using TaqMan allelic discrimination (ABI Prism® 7900HT Sequence Detection System [Applied Biosystem, Courtaboeuf, France]) according to the manufacturer recommendations as previously described [31].

2.5. Plasma methadone measures

Plasma concentration of methadone and its enantiomers, i.e., (R)‐methadone and (S)‐methadone, were measured by liquid chromatography coupled with mass spectrometry (LC/MS–MS) [32]. The analysis combined straightforward sample preparation, consisting of protein precipitation with acetonitrile, and an online enrichment by a flush/back‐flush cycle before the second‐dimension chromatography. Using D3‐deuterated internal standards allows overcoming significant relative matrix effect. This method was linear up to 2000 ng/ml. This simple sample preparation provides sensitive (the limit of quantitation is 25 ng/ml for (R,S)‐methadone. Concentration results by patient were dose adjusted (ng/ml/mg).

2.6. Statistical analyses

The statistical analyses were performed using Stata 15 software®. Deviation from the Hardy–Weinberg equilibrium was assessed using the chi‐square test. The different genotypes of patients were compared, in bivariate analyses, according to the plasma concentrations as well as opiate cessation (defined as was defined by opioid abstinence [self‐reported absence of opioid use over the previous 6 months] AND the stability or improvement of other substance use or gambling practice) by chi‐square tests or Fisher's exact tests for qualitative variables or Kruskall‐Wallis tests for quantitative variables. Then, multivariate analyses, including all genetic polymorphisms, were realized. ANOVAs were performed to explain log (R) and (S)‐methadone concentration/dose, taking into account all genotypes simultaneously. The logarithmic transformation was performed to obtain a distribution close to the Gaussian distribution. As we have already shown that CYP2B6 polymorphism (G516T; rs56308434) is related to clinical and biological response to MMT in this cohort, this variable was included in the multivariate analyses [5]. All tests were two‐tailed. The significance threshold retained was p < 0.05 and no correction for multiple testing was used according to Bender and Lange [33].

3. RESULTS

3.1. Description of the population

The present results are based on 72 patients treated with MMT for OUD. Sociodemographic and clinical characteristics of patients are resumed in Table 1. Briefly, patients were mainly men (78.0%; n = 56) and their mean age was 33.8 ± 5.7 years. The mean age for their first intake of opioids was 19.8 ± 4.5 years, 22.5 ± 5.2 years for the onset of dependence, and 25.3 ± 5.7 years for the first attempt to stop opioids. The current actual consumption of patients is presented in Table 2. Of note, among the 72 patients, 69 (95.8%) present at least one co‐addiction in addition to their OUD. Frequencies of each polymorphism studied in the 72 MMT patients are provided in Tables 3 and 4. Genotype distributions for all polymorphisms are in agreement with Hardy–Weinberg equilibrium.

TABLE 1.

Sociodemographic characteristics, history and consequences of the pathology.

Characteristic All patients (n = 72)
Men (n [%]) 56 (78%)
Age (years [m ± SD]) 33.8 ± 5.7
Graduate studies (n [%]) 29 (40%)
Marital life (n [%]) 33 (46%)
Dependent children (n [%]) 21 (29%)
Stable housing (n [%]) 61 (85%)
Professional activity (n [%]) 33 (46%)
Debt (n [%]) 29 (40%)
Consumption in the patient's circle or family (n [%]) 59 (82%)
Age at first intake (years [m ± SD]) 19.8 ± 4.5
Age at onset of dependence (years [m ± SD]) 22.4 ± 5.2
Age at the first attempt to stop (years [m ± SD]) 25.3 ± 5.7
Heroine as the principal drug (n [%]) 67 (93%)
Nasal route (n [%]) 42 (58%)
Other psychotropic drug consumption (n [%]) 60 (83%)

m: mean – SD: Standard deviation – n: number of patients.

TABLE 2.

Current actual consumption.

Characteristic All patients (n = 72)
Methadone posology (mg/day [m ± SD]) 53 ± 35
Methadone maximal posology (mg/day [m ± SD]) 80 ± 40
Good compliance (n [%]): 68 (94%)
Substance consumption (n [%]):
  • tobacco

67 (93.1%)
  • alcohol

60 (83.3%)
  • cannabis

58 (80.6%)
  • cocaine

47 (65.3%)
  • amphetamine, ecstasy

30 (41.7%)
  • LSD, NPS

28 (38.9%)
  • benzodiazepine, barbituric

30 (41.7%)
Gambling (n [%]): 33 (45.8%)
  • problematic (Lie Bet)

10

n: number of patients – LSD: Lysergic acid diethylamide – NPS: new psychoactive substances – m: mean – SD: Standard deviation – n: number of patients.

TABLE 3.

Methadone enantiomers concentrations/dose according to SNP genotypes in 72 MMT patients.

SNP Genotype n (%) (R)‐methadone/dose p‐value (S)‐methadone/dose p‐value (R,S)‐methadone/dose p‐value

ABCB1

rs1045642

TT

CT

CC

15 (21)

43 (60)

14 (19)

2.76 (1.68)

2.90 (3.03)

3.20 (1.28)

0.22

3.11 (1.84)

2.75 (2.12)

3.70 (2.06)

0.095

6.28 (3.44)

5.87 (5.15)

7.24 (3.26)

0.095

ABCB1

rs2032582

TT

TA

GG

GT

GA

13 (18)

1 (1)

20 (28)

35 (49)

3 (4)

2.61 (1.86)

2.51

3.00 (1.19)

3.08 (3.31)

2.23 (1.33)

0.59

2.78 (1.89)

1.36

3.36 (1.94)

2.96 (2.26)

2.76 (1.80)

0.50

5.83 (3.72)

3.88

3.65 (3.06)

6.29 (5.57)

5.12 (3.30)

0.53

SLC22A1

rs12208357

CC

CT

TT

57 (79)

14 (19)

1 (1)

2.95 (2.70)

2.90 (1.77)

2.30

0.94

3.04 (2.12)

2.96 (1.93)

2.14

0.78

6.27 (4.72)

6.12 (3.78)

4.66

0.82

SLC22A1

rs72552763

GAT/GAT

GAT/del

52 (72)

20 (28)

2.76 (1.31)

3.37 (4.32)

0.96

2.96 (1.71)

3.14 (2.85)

0.82

5.98 (3.03)

6.85 (7.10)

0.95

SLC22A1

rs113569197

TGGTAAGT/TGGTAAGT

TGGTAAGT/del

del/del

17 (24)

34 (47)

21 (29)

2.36 (1.05)

2.80 (1.34)

3.61 (4.22)

0.37

2.52 (1.28)

2.92 (1.37)

3.55 (3.18)

0.54

5.06 (2.42)

5.99 (2.71)

7.54 (7.21)

0.34

COMT

rs4680

GG

GA

AA

26 (36)

28 (39)

18 (25)

2.79 (1.20)

3.58 (3.72)

2.13 (0.90)

0.052

2.85 (1.98)

3.50 (2.51)

2.49 (1.09)

0.17

5.87 (3.21)

7.33 (6.24)

5.01 (1.95)

0.16

OPRM1

rs1799971

AA

AG

GG

52 (72)

19 (26)

1 (1)

2.70 (1.27)

3.63 (4.41)

1.49

0.41

2.87 (1.67)

3.49 (2.89)

1.16

0.31

5.81 (2.93)

7.52 (7.25)

2.90

0.28

n: number of patients – Kruskal‐Wallis tests were performed to compare groups – SNP: Single Nucleotide Polymorphism – Values are in ng/ml/mg.

TABLE 4.

Opiate cessation according to SNP genotypes in 72 MMT patients.

SNP Genotype Number of patients (n (%)) Opiate cessation (n (%)) p‐value

ABCB1

rs1045642

TT

CT

CC

15 (21)

43 (60)

14 (19)

8 (53.3)

30 (69.8)

10 (71.4)

0.51

ABCB1

rs2032582

TT

TA

GG

GT

GA

13 (18)

1 (1)

20 (28)

35 (49)

3 (4)

7 (53.8)

1 (100)

13 (65.0)

24 (68.6)

3 (100)

0.71

SLC22A1

rs12208357

CC

CT

TT

57 (79)

14 (19)

1 (1)

37 (64.9)

10 (71.4)

1 (100)

0.84

SLC22A1

rs72552763

GAT/GAT

GAT/del

52 (72)

20 (28)

36 (69.2)

12 (60.0)

0.58

SLC22A1

rs113569197

TGGTAAGT/TGGTAAGT

TGGTAAGT/del

del/del

17 (24)

34 (47)

21 (29)

12 (70.6)

19 (55.9)

17 (81.0)

0.16

COMT

rs4680

GG

GA

AA

26 (36)

28 (39)

18 (25)

18 (69.2)

18 (64.3)

12 (66.7)

0.95

OPRM1

rs1799971

AA

AG

GG

52 (72)

19 (26)

1 (1)

35 (67.3)

13 (68.4)

0 (0.0)

0.51

n: number of patients – Fisher's exact tests were performed to compare groups – SNP: Single Nucleotide Polymorphism.

3.2. Influence of genotype on dose‐adjusted plasma methadone levels

Bivariate analyses for codominant models are presented in Table 3. In these codominant models, levels of (R,S)‐methadone/dose, of (R)‐methadone/dose, and of (S)‐methadone/dose were not significantly different for all polymorphisms tested of ABCB1, SLC22A1, COMT or OPRM1 (Table 3).

Recessive models were also performed for all polymorphisms. For rs1045642 of ABCB1 (TT vs. CT + CC), no significant difference was evidenced between groups for (R,S)‐methadone/dose (6.28 ± 3.44 vs. 6.21 ± 4.76 ng/ml/mg; p = 0.78), for (R)‐methadone/dose (2.76 ± 1.68 vs. 2.98 ± 2.70 ng/ml/mg; p = 0.64), nor for (S)‐methadone/dose (3.11 ± 1.84 vs. 2.98 ± 2.13 ng/ml/mg; p = 0.78). As well, for rs2032582 of ABCB1 (TT + TA + AA vs. GG + GT + GA), no significant difference was evidenced between groups for (R,S)‐methadone/dose (5.69 ± 3.61 vs. 6.35 ± 4.71 ng/ml/mg; p = 0.37), for (R)‐methadone/dose (2.60 ± 1.78 vs. 3.01 ± 2.66 ng/ml/mg; p = 0.30), nor for (S)‐methadone/dose (2.68 ± 1.86 vs. 3.09 ± 2.11 ng/ml/mg; p = 0.29).

Regarding rs12208357 (CC vs. CT + TT) and rs113569197 (TGGTAAGT/TGGTAAGT vs. TGGTAAGT/del + del/del) of SLC22A1, there was no significant difference between groups for (R,S)‐methadone/dose (respectively, 6.27 ± 4.72 vs. 6.02 ± 3.66 ng/ml/mg; p = 0.66 and 5.06 ± 2.42 vs. 6.58 ± 4.93 ng/ml/mg; p = 0.14), for (R)‐methadone/dose (respectively, 2.95 ± 2.70 vs. 2.86 ± 1.71 ng/ml/mg; p = 0.88 and 2.36 ± 1.05 vs. 3.11 ± 2.80 ng/ml/mg; p = 0.19), nor for (S)‐methadone/dose (respectively, 3.04 ± 2.12 vs. 2.90 ± 1.87 ng/ml/mg; p = 0.57 and 2.52 ± 1.28 vs. 3.16 ± 2.23 ng/ml/mg; p = 0.27).

Concerning rs4680 (GG vs. GA + AA) of COMT, no significant difference was found between groups for (R,S)‐methadone/dose, (R)‐methadone/dose and (S)‐methadone/dose (respectively, 5.87 ± 3.21 vs. 6.42 ± 5.11 ng/ml/mg; p = 0.53, 2.79 ± 1.20 vs. 3.01 ± 3.02 ng/ml/mg; p = 0.66 and 2.85 ± 1.98 vs. 3.10 ± 2.12 ng/ml/mg; p = 0.32).

Finally, with regard to rs1799971 (AA vs. AG + GG) of OPRM1, no difference was also identified between groups (R,S)‐methadone/dose, (R)‐methadone/dose and (S)‐methadone/dose (respectively, 5.81 ± 2.93 vs. 7.29 ± 7.13 ng/ml/mg; p = 0.66, 2.70 ± 1.27 vs. 3.52 ± 4.32 ng/ml/mg; p = 0.89 and 2.87 ± 1.67 vs. 3.37 ± 2.86 ng/ml/mg; p = 0.79).

In multivariate analyses, none of the polymorphisms explained the (R)‐methadone concentration/dose values. Nevertheless, in a stepwise elimination process, the latest eliminated variable was rs4680 of COMT (p = 0.054). Also, for the (S)‐methadone concentration/dose values, there was no association identified (except for rs56308434 of CYP2B6; p = 0.003). In the stepwise elimination process, the latest eliminated variable was rs1045642 of ABCB1 (p = 0.11). Finally, no association was identified between studied polymorphisms and the (R,S)‐methadone concentration/dose values (except for rs56308434 of CYP2B6; p = 0.023). However, it should be noted that in a stepwise elimination process, the latest eliminated variable was rs113569197 of SLC22A1 (p = 0.060).

3.3. Influence of genotype on opiate cessation

Bivariate analyses for codominant models are presented in Table 4. In these codominant models, opiate abstinence during treatment by methadone was not associated with all polymorphisms tested of ABCB1, SLC22A1, COMT, or OPRM1 (Table 4).

As for the biological parameters, recessive models were performed for opiate abstinence, as previously described. For ABCB1, both polymorphisms (rs1045642 and rs2032582) were not associated with opiate abstinence (respectively, 53.3% vs. 70.2%; p = 0.23, and 57.1% vs. 69.0%; p = 0.53).

For SLC22A1, no significant difference between groups was evidenced for rs12208357 and rs113569197 (respectively, 64.9% vs. 73.3%; p = 0.39, and 70.6% vs. 65.5%; p = 0.78).

Finally, for the pharmacodynamic targets, rs4680 of COMT and rs1799971 of OPRM1 were not associated with opiate abstinence in our sample during MMT (respectively, 69.2% vs. 65.2%; p = 0.80, and 67.3% vs. 65.0%; p = 0.53).

4. DISCUSSION

This study assessed associations between various genetic polymorphisms from ABCB1, SLC22A1, COMT, and OPRM1 with clinical response to MMT and pharmacological characteristics of patients with OUD receiving MMT. We did not find any statistically significant association between these polymorphisms and clinical outcomes (opiate cessation). Moreover, we did not detect any association between dose‐adjusted plasma methadone levels and these genetic variants.

We explored pharmacogenetics variants either pharmacokinetic (ABCB1 and SLC22A1) or pharmacodynamic (COMT and OPRM1) targets. Pharmacokinetic variants affect the bioavailability of the medication in the body, while pharmacodynamic variants directly or indirectly alter the effects of the medication. Among the pharmacokinetic targets, we previously showed that genetic polymorphisms of enzyme proteins such as CYP2B6 could be associated with pharmacological parameters and clinical responses to MMT5. Nevertheless, non‐enzyme proteins can also affect methadone bioavailability; methadone is a substrate of the P‐glycoprotein efflux transporter which is encoded by ABCB1. ABCB1 polymorphisms can influence the transport of opioids at the intestinal level and at the blood–brain barrier [18, 34, 35]. In our study, neither rs1045642 nor rs2032582 of ABCB1 are associated with dose‐adjusted plasma methadone concentrations. These results are globally in concordance with previous research. Indeed, most studies investigating the impact of ABCB1 polymorphisms on methadone response have reported associations with prescribed methadone doses [12, 28, 36], while generally observing no significant association with plasma methadone concentrations, with some exceptions noted [14, 16, 27]. In addition, another publication identified associations between rs1045642 and brain/blood ratios of methadone in methadone‐related deaths [37]. These results suggest that the impact of ABCB1 polymorphisms on methadone pharmacokinetic parameters and methadone responses is related to their effects methadone diffusion into the brain across the blood–brain barrier rather than to their effects on plasma concentrations of methadone [37]. This hypothesis has been tested and validated in rodent models [38].

The second pharmacokinetics target tested is OCT1 (coded by SLC22A1). Recently, loss‐of‐function polymorphisms of SLC22A1 have been reported to be associated with reduced postoperative tramadol consumption [39]. Moreover, loss‐of‐function genetic variants of this gene were associated with higher plasma concentrations of O‐desmethyltramadol and morphine in patients and in healthy subjects [40, 41]. Implication of these polymorphisms for methadone has not been yet evidenced, and to the best of our knowledge, we are the first ones which assess the association between SLC22A1 polymorphisms and clinical/biological responses to MMT and found that there was no relation between them. To date, these results are not unexpected, as methadone does not appear to be a direct substrate of OCT1 [42]. Nevertheless, the influence of SLC22A1 polymorphisms could be indirectly relevant. First, OCT1 is one of the main transporters of EDDP (2‐ethylidene‐1,5‐dimethyl‐3,3‐diphenylpyrrolidine), a primary metabolite of methadone [42]. Thus, an alteration in the transport of EDDP could modify methadone metabolism. Second, OCT1 serves as a primary transporter of acylcarnitines, which have been associated with methadone withdrawal syndrome. This observation could suggest an indirect pathway by which acylcarnitines, potentially mediated by OCT1, may influence methadone management—a connection already observed in depression [43, 44, 45].

Concerning the pharmacodynamic targets, COMT is the enzyme that realizes the methyl conjugation of catecholamines such as dopamine and is, consequently, a key regulator of their concentrations [23]. In the context of addiction, the dopaminergic system was explored given that this pathway explained a part of the rewarding and reinforcing effects of drugs [46]. The most studied polymorphism of COMT is rs4880 and in vitro data suggest that it produces a protein with a 3–4‐fold lower enzymatic activity [23, 47]. As expected, this polymorphism was not associated with dose‐adjusted plasma methadone concentrations, since it is involved in the pharmacokinetic mechanisms of methadone. However, it was not associated with opiate abstinence during MMT in patients. Our data are consistent with previous studies. Indeed, in other cohorts from 81 to 820 patients, this polymorphism was not related to methadone prescribed dose, clinical response during MMT, or adverse events [23, 28, 48]. Only another variant of COMT, rs933271, was described to be associated with opiate cessation in a Han Chinese population [48].

The second studied pharmacodynamic gene is OPRM1, coding for the μ‐opioid receptor which is one of the main targets of methadone for inducing its pharmacological effect making this receptor a major candidate to explain genetic inter‐individual differences in opioid response [49]. The studied polymorphism, rs1799971, is one of the most common genetic variants of OPRM1 and is known to induce a reduced response to opioids [49]. This effect could be due to the removal of a glycosylation site in the N‐terminal domain of the receptor and a reduction in gene expression [49, 50]. This genetic variant was described to be associated with the response to other opioid drugs such as naltrexone or buprenorphine [51, 52]. It was expected that there would be no significant difference in dose‐adjusted plasma methadone concentrations according to the rs1799971 OPRM1 genetic polymorphism. But we equally did not highlight any difference for opiate cessation during MMT according to this last polymorphism. The effect of this polymorphism remains highly controversial both in terms of methadone prescribed dose and therapeutic efficacy [12, 23, 28, 53, 54, 55, 56]. Nevertheless, in concordance with our observed result, an in vitro study suggests that this genetic variant does not alter methadone signaling in a cellular model [52].

This study had some limitations. First, the sample size is relatively small. But this size allowed a great phenotyping of patients with a wealth of clinical data at our disposal. Second, we assessed only methadone whereas other medications are available such as buprenorphine to manage OUD. But this approach reinforces the biological rationale. Third, except for CYP2B6 polymorphisms, no other cofounding factors, such co‐medications or potential hepatic dysfunction, were included in the multivariate analyses as these data were not available. Nonetheless, in clinical practice, few drugs with pharmacokinetic drug–drug interactions are prescribed, given the limited number associated with CYP2B6 induction or inhibition within the pharmacopeia [57]. Similarly, recent data suggest that liver diseases do not significantly influence the pharmacokinetic parameters of methadone [58]. Fourth, we did not perform urine tests to screen substance use in addition to declarative status. We may assume that the unsuccessful OUD treatment rate could be underestimated. Finally , genetic polymorphisms were analyzed separately while some data suggest that the therapeutic effect could be observed with a combination of genetic variants [12, 35, 59]. Hence, their combination could be associated with response to MMT. In accordance, a genetic test that combines several polymorphisms (including ABCB1, COMT, and OPRM1 variants) has been recently approved by the US Food And Drug Administration (FDA) to estimate the risk of developing OUD [59, 60]. But our sample size was not sufficient to perform this type of analysis. Nonetheless, cohorts with a large number of subjects allowing the identification of polygenic biomarkers are rarely available in pharmacogenetic studies, particularly in the context of OUD5. Similarly, the selection of polymorphisms was based on their frequency and the availability of previous data, such as their association with the response to other treatments which remains hypothetical. Consequently, we cannot exclude the possibility that other variants not studied here, particularly rare ones, may have an impact on the phenotypes studied. However, the size of our cohort did not allow us to adopt an alternative approach that would have included them. Of note, the prescribed methadone dosages were relatively low, which may be inherent to the naturalistic approach used in our real‐world study. Furthermore, it is unlikely that this would influence the associations being investigated between concentrations and genetic polymorphisms of the targets, as concentrations were normalized to the received dosage.

5. CONCLUSION

To conclude, in our sample, all the polymorphisms tested in these genes were not associated with clinical responses or pharmacological parameters to methadone. Hence, ABCB1, SLC22A1, COMT, and OPRM1 genotyping cannot yet be considered in a daily clinical setting for the management of MMT in OUD. Further investigations in larger cohorts, exploring other polymorphisms or targets and combining various polymorphisms should be performed to confirm or refute these preliminary findings.

AUTHORS CONTRIBUTION

Abd El Kader Ait Tayeb: Writing—original draft/formal analysis; visualization; resources; validation; writing—review & editing. Edouard‐Jules Laforgue: Investigation; data curation; writing—review & editing. Benoit Schreck: Investigation; data curation; writing—review & editing. Marie Grall‐Bronnec: Conceptualization; investigation; data curation; writing—review & editing. Jean‐Benoit Hardouin: Formal analysis; writing—review & editing. Juliette Leboucher: Conceptualization; writing—review & editing. Caroline Victorri‐Vigneau: Funding acquisition; supervision; methodology; validation; writing—review & editing. Céline Verstuyft: Supervision; methodology; resources; validation; writing—review & editing.

CONFLICT OF INTEREST STATEMENT

Abd El Kader Ait Tayeb, Edouard‐Jules Laforgue, Benoit Schreck, Marie Grall‐Bronnec, Jean‐Benoit Hardouin, Juliette Leboucher, Caroline Victorri‐Vigneau, Céline Verstuyft have no conflict of interest to disclose.

ACKNOWLEDGMENTS

This study was supported jointly by the Mission Interministérielle de Lutte contre les Drogues et les Conduites Addictives (MILDECA) and the Université Paris 13, as part of the call for research projects “PREVDROG” launched by these two organizations in 2011. It was conducted at the initiative of and coordinated by the UIC “Psychiatrie et Santé Mentale” of Nantes University Hospital. Nantes University Hospital is the sponsor of this study.

Ait Tayeb AEK, Laforgue E‐J, Schreck B, et al. ABCB1, SLC22A1, COMT, and OPRM1 genotypes: Study of their influence on plasma methadone levels and clinical response to methadone maintenance treatment in opioid use disorder. Fundam Clin Pharmacol. 2025;39(3):e70013. doi: 10.1111/fcp.70013

Contributor Information

Abd El Kader Ait Tayeb, Email: abdelkader.aittayeb@aphp.fr.

OPAL Group:

Caroline Victorri‐Vigneau and Céline Verstuyft

DATA AVAILABILITY STATEMENT

The datasets generated and analyzed during the current study are not publicly available because the data generated included sensitive data according to the French Data Protection Authority (CNIL), that could not be transferred to other researchers to guarantee participants' anonymity. But they are available from the corresponding author on reasonable request.

REFERENCES

  • 1. Degenhardt L, Grebely J, Stone J, et al. Global patterns of opioid use and dependence: harms to populations, interventions, and future action. Lancet. 2019;394(10208):1560‐1579. doi: 10.1016/S0140-6736(19)32229-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Blanco C, Volkow ND. Management of opioid use disorder in the USA: present status and future directions. Lancet. 2019;393(10182):1760‐1772. doi: 10.1016/S0140-6736(18)33078-2 [DOI] [PubMed] [Google Scholar]
  • 3. Suarez EA, Huybrechts KF, Straub L, et al. Buprenorphine versus methadone for opioid use disorder in pregnancy. N Engl J Med. 2022;387(22):2033‐2044. doi: 10.1056/NEJMoa2203318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Haute Autorité de Santé (HAS) , Bon usage des médicaments opioïdes: antalgie, prévention et prise en charge du trouble de l'usage et des surdoses, 2022.
  • 5. Victorri‐Vigneau C, Verstuyft C, Bouquié R, et al. Relevance of CYP2B6 and CYP2D6 genotypes to methadone pharmacokinetics and response in the OPAL study. Br J Clin Pharmacol. 2019;85(7):1538‐1543. doi: 10.1111/bcp.13936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Crettol S, Deglon J, Besson J, et al. Methadone enantiomer plasma levels, CYP2B6, CYP2C19, and CYP2C9 genotypes, and response to treatment. Clin Pharm Therap. 2005;78(6):593‐604. doi: 10.1016/j.clpt.2005.08.011 [DOI] [PubMed] [Google Scholar]
  • 7. Ansermot N. Substitution of (R,S)‐methadone by (R)‐methadone: impact on QTc interval. Arch Intern Med. 2010;170(6):529. doi: 10.1001/archinternmed.2010.26 [DOI] [PubMed] [Google Scholar]
  • 8. Ahmad T, Valentovic MA, Rankin GO. Effects of cytochrome P450 single nucleotide polymorphisms on methadone metabolism and pharmacodynamics. Biochem Pharmacol. 2018;153:196‐204. doi: 10.1016/j.bcp.2018.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Csajka C, Crettol S, Guidi M, Eap CB. Population genetic‐based pharmacokinetic modeling of methadone and its relationship with the QTc interval in opioid‐dependent patients. Clin Pharmacokinet. 2016;55(12):1521‐1533. doi: 10.1007/s40262-016-0415-2 [DOI] [PubMed] [Google Scholar]
  • 10. Eap CB, Bourquin M, Martin JL, et al. Plasma concentrations of the enantiomers of methadone and therapeutic response in methadone maintenance treatment. Drug Alcohol Depend. 2000;61(1):47‐54. doi: 10.1016/S0376-8716(00)00121-6 [DOI] [PubMed] [Google Scholar]
  • 11. Crettol S, Digon P, Powell Golay K, Brawand M, Eap CB. In vitro P‐glycoprotein‐mediated transport of (R)‐, (S)‐, (R,S)‐methadone, LAAM and their main metabolites. Pharmacology. 2007;80(4):304‐311. doi: 10.1159/000107104 [DOI] [PubMed] [Google Scholar]
  • 12. Hung CC, Chiou MH, Huang BH, et al. Impact of genetic polymorphisms in ABCB1, CYP2B6, OPRM1, ANKK1 and DRD2 genes on methadone therapy in Han Chinese patients. Pharmacogenomics. 2011;12(11):1525‐1533. doi: 10.2217/pgs.11.96 [DOI] [PubMed] [Google Scholar]
  • 13. Dobrinas M, Crettol S, Oneda B, et al. Contribution of CYP2B6 alleles in explaining extreme (S)‐methadone plasma levels: a CYP2B6 gene resequencing study. Pharmacogenet Genomics. 2013;23(2):84‐93. doi: 10.1097/FPC.0b013e32835cb2e2 [DOI] [PubMed] [Google Scholar]
  • 14. Crettol S, Deglon J, Besson J, et al. ABCB1 and cytochrome P450 genotypes and phenotypes: influence on methadone plasma levels and response to treatment. Clin Pharm Therap. 2006;80(6):668‐681. doi: 10.1016/j.clpt.2006.09.012 [DOI] [PubMed] [Google Scholar]
  • 15. Benmebarek M. Effects of grapefruit juice on the pharmacokinetics of the enantiomers of methadone*1. Clin Pharm Therap. 2004;76(1):55‐63. doi: 10.1016/j.clpt.2004.03.007 [DOI] [PubMed] [Google Scholar]
  • 16. Zahari Z, Lee CS, Ibrahim MA, et al. Relationship between ABCB1 polymorphisms and serum methadone concentration in patients undergoing methadone maintenance therapy (MMT). Am J Drug Alcohol Abuse. 2016;42(5):587‐596. doi: 10.3109/00952990.2016.1172078 [DOI] [PubMed] [Google Scholar]
  • 17. Eap CB, Broly F, Mino A, et al. Cytochrome P450 2D6 genotype and methadone steady‐state concentrations. J Clin Psychopharmacol. 2001;21(2):229‐234. doi: 10.1097/00004714-200104000-00016 [DOI] [PubMed] [Google Scholar]
  • 18. Matic M, de Wildt SN, Tibboel D, van Schaik RHN. Analgesia and opioids: a pharmacogenetics shortlist for implementation in clinical practice. Clin Chem. 2017;63(7):1204‐1213. doi: 10.1373/clinchem.2016.264986 [DOI] [PubMed] [Google Scholar]
  • 19. AlMeman A, Ismail R, Perola M. CYP2B6 and OPRM1 receptor polymorphisms at methadone clinics and novel OPRM1 haplotypes: a cross‐sectional study. DML. 2016;10(3):213‐218. doi: 10.2174/1872312810666160810104040 [DOI] [PubMed] [Google Scholar]
  • 20. Crist RC, Doyle GA, Nelson EC, et al. A polymorphism in the OPRM1 3′‐untranslated region is associated with methadone efficacy in treating opioid dependence. Pharmacogenomics J. 2018;18(1):173‐179. doi: 10.1038/tpj.2016.89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Oneda B, Crettol S, Bochud M, et al. β‐Arrestin2 influences the response to methadone in opioid‐dependent patients. Pharmacogenomics J. 2011;11(4):258‐266. doi: 10.1038/tpj.2010.37 [DOI] [PubMed] [Google Scholar]
  • 22. Crettol S, Besson J, Croquette‐Krokar M, et al. Association of dopamine and opioid receptor genetic polymorphisms with response to methadone maintenance treatment. Prog Neuro‐Psychopharmacol Biol Psychiatry. 2008;32(7):1722‐1727. doi: 10.1016/j.pnpbp.2008.07.009 [DOI] [PubMed] [Google Scholar]
  • 23. Crews KR, Monte AA, Huddart R, et al. Clinical pharmacogenetics implementation consortium guideline for CYP2D6, OPRM1, and COMT genotypes and select opioid therapy. Clin Pharm Therap. 2021;110(4):888‐896. doi: 10.1002/cpt.2149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Somogyi AA, Barratt DT, Ali RL, Coller JK. Pharmacogenomics of methadone maintenance treatment. Pharmacogenomics. 2014;15(7):1007‐1027. doi: 10.2217/pgs.14.56 [DOI] [PubMed] [Google Scholar]
  • 25. Yang HC, Chu SK, Huang CL, et al. Genome‐wide Pharmacogenomic study on methadone maintenance treatment identifies SNP rs17180299 and multiple haplotypes on CYP2B6, SPON1, and GSG1L associated with plasma concentrations of methadone R‐ and S‐enantiomers in heroin‐dependent patients. Barsh GS, ed. PLoS Genet. 2016;12(3):e1005910. doi: 10.1371/journal.pgen.1005910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Marie‐Claire C, Crettol S, Cagnard N, et al. Variability of response to methadone: genome‐wide DNA methylation analysis in two independent cohorts. Epigenomics. 2016;8(2):181‐195. doi: 10.2217/epi.15.110 [DOI] [PubMed] [Google Scholar]
  • 27. Fonseca F, de la Torre R, Díaz L, Pastor A, Cuyàs E, Pizarro N, Khymenets O, Farré M, Torrens M Contribution of cytochrome P450 and ABCB1 genetic variability on methadone pharmacokinetics, dose requirements, and response. Zanger UM, ed. PLoS ONE. 2011;6(5):e19527. 10.1371/journal.pone.0019527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Mouly S, Bloch V, Peoc'h K, et al. Methadone dose in heroin‐dependent patients: role of clinical factors, comedications, genetic polymorphisms and enzyme activity. Brit J Clinical Pharm. 2015;79(6):967‐977. doi: 10.1111/bcp.12576 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Grall‐Bronnec M, Laforgue EJ, Challet‐Bouju G, et al. Prevalence of coaddictions and rate of successful treatment among a French sample of opioid‐dependent patients with long‐term opioid substitution therapy: the OPAL study. Front Psych. 2019;10:726. doi: 10.3389/fpsyt.2019.00726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Whirl‐Carrillo M, Huddart R, Gong L, et al. An evidence‐based framework for evaluating pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2021;110(3):563‐572. doi: 10.1002/cpt.2350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Coulbault L, Beaussier M, Verstuyft C, et al. Environmental and genetic factors associated with morphine response in the postoperative period. Clin Pharmacol Therap. 2006;79(4):316‐324. doi: 10.1016/j.clpt.2006.01.007 [DOI] [PubMed] [Google Scholar]
  • 32. Bouquié R, Hernando H, Deslandes G, et al. Chiral on‐line solid phase extraction coupled to liquid chromatography–tandem mass spectrometry assay for quantification of (R) and (S) enantiomers of methadone and its main metabolite in plasma. Talanta. 2015;134:373‐378. doi: 10.1016/j.talanta.2014.11.052 [DOI] [PubMed] [Google Scholar]
  • 33. Bender R, Lange S. Adjusting for multiple testing—when and how? J Clin Epidemiol. 2001;54(4):343‐349. doi: 10.1016/S0895-4356(00)00314-0 [DOI] [PubMed] [Google Scholar]
  • 34. Baber M, Bapat P, Nichol G, Koren G. The pharmacogenetics of opioid therapy in the management of postpartum pain: a systematic review. Pharmacogenomics. 2016;17(1):75‐93. doi: 10.2217/pgs.15.157 [DOI] [PubMed] [Google Scholar]
  • 35. Bell GC, Donovan KA, McLeod HL. Clinical implications of opioid pharmacogenomics in patients with cancer. Cancer Control. 2015;22(4):426‐432. doi: 10.1177/107327481502200408 [DOI] [PubMed] [Google Scholar]
  • 36. Levran O, O'Hara K, Peles E, et al. ABCB1 (MDR1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Hum Mol Genet. 2008;17(14):2219‐2227. doi: 10.1093/hmg/ddn122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. 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]
  • 38. 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. 2004;173(1–2):132‐138. doi: 10.1007/s00213-003-1718-1 [DOI] [PubMed] [Google Scholar]
  • 39. Stamer UM, Musshoff F, Stüber F, Brockmöller J, Steffens M, Tzvetkov MV. Loss‐of‐function polymorphisms in the organic cation transporter OCT1 are associated with reduced postoperative tramadol consumption. Pain. 2016;157(11):2467‐2475. doi: 10.1097/j.pain.0000000000000662 [DOI] [PubMed] [Google Scholar]
  • 40. Tzvetkov MV, dos Santos Pereira JN, Meineke I, Saadatmand AR, Stingl JC, Brockmöller J. Morphine is a substrate of the organic cation transporter OCT1 and polymorphisms in OCT1 gene affect morphine pharmacokinetics after codeine administration. Biochem Pharmacol. 2013;86(5):666‐678. doi: 10.1016/j.bcp.2013.06.019 [DOI] [PubMed] [Google Scholar]
  • 41. Tzvetkov MV, Saadatmand AR, Lötsch J, Tegeder I, Stingl JC, Brockmöller J. Genetically polymorphic OCT1: another piece in the puzzle of the variable pharmacokinetics and pharmacodynamics of the opioidergic drug tramadol. Clin Pharmacol Ther. 2011;90(1):143‐150. doi: 10.1038/clpt.2011.56 [DOI] [PubMed] [Google Scholar]
  • 42. Campbell SD, Gadel S, Friedel C, Crafford A, Regina KJ, Kharasch ED. Influence of HIV antiretrovirals on methadone N‐demethylation and transport. Biochem Pharmacol. 2015;95(2):115‐125. doi: 10.1016/j.bcp.2015.03.007 [DOI] [PubMed] [Google Scholar]
  • 43. Ait Tayeb AEK, Colle R, El‐Asmar K, et al. Plasma acetyl‐ l ‐carnitine and l ‐carnitine in major depressive episodes: a case–control study before and after treatment. Psychol Med. 2021;14(6):1‐10. doi: 10.1017/S003329172100413X [DOI] [PubMed] [Google Scholar]
  • 44. Janiri L, Martinotti G, Tonioni F, et al. Acetyl‐L‐carnitine in the management of pain during methadone withdrawal syndrome. Clin Neuropharmacol. 2009;32(1):35‐40. doi: 10.1097/WNF.0b013e31815dd465 [DOI] [PubMed] [Google Scholar]
  • 45. Matthaei J, Brockmöller J, Steimer W, et al. Effects of genetic polymorphism in CYP2D6, CYP2C19, and the organic cation transporter OCT1 on amitriptyline pharmacokinetics in healthy volunteers and depressive disorder patients. Front Pharmacol. 2021;12:688950. doi: 10.3389/fphar.2021.688950 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Volkow ND, Boyle M. Neuroscience of addiction: relevance to prevention and treatment. AJP. 2018;175(8):729‐740. doi: 10.1176/appi.ajp.2018.17101174 [DOI] [PubMed] [Google Scholar]
  • 47. Taranu A, Asmar KE, Colle R, et al. The catechol‐O‐methyltransferase Val(108/158)met genetic polymorphism cannot be recommended as a biomarker for the prediction of venlafaxine efficacy in patients treated in psychiatric settings. Basic Clin Pharmacol Toxicol. 2017;121(5):435‐441. doi: 10.1111/bcpt.12827 [DOI] [PubMed] [Google Scholar]
  • 48. Duan L, Li X, Yan J, et al. Association of COMT gene polymorphisms with response to methadone maintenance treatment among Chinese opioid‐dependent patients. Genet Test Mol Biomarkers. 2020;24(6):364‐369. doi: 10.1089/gtmb.2019.0275 [DOI] [PubMed] [Google Scholar]
  • 49. Knapman A, Connor M. Cellular signalling of non‐synonymous single‐nucleotide polymorphisms of the human μ‐opioid receptor (OPRM1). Br J Pharmacol. 2015;172(2):349‐363. doi: 10.1111/bph.12644 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Zhang Y, Wang D, Johnson AD, Papp AC, Sadée W. Allelic expression imbalance of human mu opioid receptor (OPRM1) caused by variant A118G. J Biol Chem. 2005;280(38):32618‐32624. doi: 10.1074/jbc.M504942200 [DOI] [PubMed] [Google Scholar]
  • 51. Verholleman A, Victorri‐Vigneau C, Laforgue E, Derkinderen P, Verstuyft C, Grall‐Bronnec M. Naltrexone use in treating Hypersexuality induced by dopamine replacement therapy: impact of OPRM1 A/G polymorphism on its effectiveness. IJMS. 2020;21(8):3002. doi: 10.3390/ijms21083002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Knapman A, Santiago M, Connor M. Buprenorphine signalling is compromised at the n 40 d polymorphism of the human μ opioid receptor in vitro . Br J Pharm. 2014;171(18):4273‐4288. doi: 10.1111/bph.12785 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Kazi I, Chenoweth MJ, Jutras‐Aswad D, et al. Pharmacogenetics of biochemically verified abstinence in an opioid agonist therapy randomized clinical trial of methadone and buprenorphine/naloxone. Clin Pharm Therap. Published online December 6. 2023;cpt.3112. doi: 10.1002/cpt.3112 [DOI] [PubMed] [Google Scholar]
  • 54. Levran O, Peles E, Randesi M, da Rosa JC, Adelson M, Kreek MJ. The μ‐opioid receptor nonsynonymous variant 118A>G is associated with prolonged abstinence from heroin without agonist treatment. Pharmacogenomics. 2017;18(15):1387‐1391. doi: 10.2217/pgs-2017-0092 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Takemura M, Niki K, Okamoto Y, et al. Comparison of the effects of OPRM1 A118G polymorphism using different opioids: a prospective study. J Pain Symptom Manag. Published online September. 2023;S0885392423006875. doi: 10.1016/j.jpainsymman.2023.09.017 [DOI] [PubMed] [Google Scholar]
  • 56. Xie X, Gu J, Zhuang D, et al. Association between rs1799971 in the mu opioid receptor gene and methadone maintenance treatment response. Clin Lab Anal. 2022;36(11):e24750. doi: 10.1002/jcla.24750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Miano TA, Wang L, Leonard CE, et al. Identifying clinically relevant drug–drug interactions with methadone and buprenorphine: a translational approach to signal detection. Clin Pharm Therap. 2022;112(5):1120‐1129. doi: 10.1002/cpt.2717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Chalabianloo F, Fadnes LT, Johansson KA, et al. Methadone pharmacokinetics in opioid agonist treatment: influencing factors and clinical implications. Basic Clin Pharmacol Toxicol. 2024;134(3):333‐344. doi: 10.1111/bcpt.13975 [DOI] [PubMed] [Google Scholar]
  • 59. Donaldson K, Cardamone D, Genovese M, Garbely J, Demers L. Clinical performance of a gene‐based machine learning classifier in assessing risk of developing OUD in subjects taking oral opioids: a prospective observational study. Ann Clin Lab Sci. 2021;51(4):451‐460. [PubMed] [Google Scholar]
  • 60.Available from: https://www.fda.gov/medical-devices/medical-devices-news-and-events/fda-approves-first-test-help-identify-elevated-risk-developing-opioid-use-disorder

Associated Data

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

Data Availability Statement

The datasets generated and analyzed during the current study are not publicly available because the data generated included sensitive data according to the French Data Protection Authority (CNIL), that could not be transferred to other researchers to guarantee participants' anonymity. But they are available from the corresponding author on reasonable request.


Articles from Fundamental & Clinical Pharmacology are provided here courtesy of Wiley

RESOURCES