* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Methadone—metabolism, pharmacokinetics and interactions
Discovery and development of cyclooxygenase 2 inhibitors wikipedia , lookup
Polysubstance dependence wikipedia , lookup
Drug design wikipedia , lookup
Plateau principle wikipedia , lookup
Neuropsychopharmacology wikipedia , lookup
Drug discovery wikipedia , lookup
Theralizumab wikipedia , lookup
Prescription drug prices in the United States wikipedia , lookup
Pharmaceutical industry wikipedia , lookup
Psychopharmacology wikipedia , lookup
Pharmacognosy wikipedia , lookup
Neuropharmacology wikipedia , lookup
Prescription costs wikipedia , lookup
Dydrogesterone wikipedia , lookup
Pharmacogenomics wikipedia , lookup
Pharmacological Research 50 (2004) 551–559 Methadone—metabolism, pharmacokinetics and interactions Anna Ferrari∗ , Ciro Pio Rosario Coccia, Alfio Bertolini, Emilio Sternieri Section of Toxicology and Clinical Pharmacology, University of Modena and Reggio Emilia, Policlinico, Largo del Pozzo, 71-41100 Modena, Italy Accepted 4 May 2004 Abstract The pharmacokinetics of methadone varies greatly from person to person; so, after the administration of the same dose, considerably different concentrations are obtained in different subjects, and the pharmacological effect may be too small in some patients, too strong and prolonged in others. Methadone is mostly metabolised in the liver; the main step consists in the N-demethylation by CYP3A4 to EDDP (2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine), an inactive metabolite. The activity of CYP3A4 varies considerably among individuals, and such variability is the responsible for the large differences in methadone bioavailability. CYP2D6 and probably CYP1A2 are also involved in methadone metabolism. During maintenance treatment with methadone, treatment with other drugs may be necessary due to the frequent comorbidity of drug addicts: psychotropic drugs, antibiotics, anticonvulsants and antiretroviral drugs, which can cause pharmacokinetic interactions. In particular, antiretrovirals, which are CYP3A4 inducers, can decrease the levels of methadone, so causing withdrawal symptoms. Buprenorphine, too, is metabolised by CYP3A4, and may undergo the same interactions as methadone. Since it is impossible to foresee the time-lapse from the administration of another drug to the appearing of withdrawal symptoms, nor how much the daily dose of methadone should be increased in order to prevent them, patients taking combined drug treatments must be carefully monitored. The so far known pharmacokinetic drug–drug interactions of methadone do not have life-threatening consequences for the patients, but they usually cause a decrease of the concentrations and of the effects of the drug, which in turn can cause symptoms of withdrawal and increase the risk of relapse into heroin abuse. © 2004 Elsevier Ltd. All rights reserved. Keywords: Methadone; Drug–drug interactions; Pharmacokinetic; CYP3A4; CYP2D6 1. Introduction Since 1965, the year in which Dole and Nyswander [1] proposed the introduction of methadone as a substitute for heroin, its use has spread progressively also in Italy, in particular for the treatment of drug addicts who cannot remain drug-free in spite of detoxication therapies and attendance in therapeutic communities. Maintenance treatment with methadone, performed with doses adequate to the actual needs of the individual addict, contributes to a drop in mortality, to stopping or reducing heroin use, to decreasing or avoiding relapses and criminal activity, to favouring the finding of a job and improving family and social relationships, to reducing the risk of HIV and hepatitis virus infections [2]. The pharmacological characteristics that support the use of methadone as a replacement in the long term treatment ∗ Corresponding author. Tel.: +39 59 4224064; fax: +39 59 4224069. E-mail address: [email protected] (A. Ferrari). 1043-6618/$ – see front matter © 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.phrs.2004.05.002 of heroin addiction, a pathological condition that has been defined as a “chronic relapsing disorder” [3], are the high oral bioavailability, the long elimination time that makes a single daily administration possible, the lack of behavioural modifications such as to be detrimental to persons carrying out normal work activities, and the availability of a specific antagonist that can be used in the case of overdose. The most negative kinetic characteristics are the inter-individual variability of absorption and metabolism [4] which make it impossible to anticipate, with acceptable approximation, the relationship between dose, blood concentration, and clinical effect [3]. 2. Pharmacokinetics of methadone The available methadone hydrochloride on the market is a racemic mixture of two stereoisomers. l-Methadone is the pharmacologically active isomer [5–7] (however, d-methadone retains certain pharmacological effects; for example, the antitussive activity.) Methadone taken orally is 552 A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 subjected to an important first-pass effect and is detectable in the plasma about 30 min after administration [8]. Its bioavailability varies from 41–76 [9] to 85–95% [10]. Thus, following the administration of equal doses, quite different blood concentrations are obtained in different subjects [4,11]. This variability must be kept in mind when selecting the initial doses of methadone, and in settling the optimum dosages for maintenance treatment. There are considerable differences also in the time needed to reach the maximum plasma concentration (Tmax ): from 1 to 6 h, with average values of 2.5–4.4 h [9,10,12]. The course of methadone plasma concentrations over time follows a bi-exponential curve, with a rapid ␣-phase, that corresponds to the transfer of the drug from the central compartment to the tissue compartment and to the beginning of elimination, and a slow -phase, that corresponds to elimination [13]. The t1/2 of the first phase of methadone disappearance from the plasma (␣-phase, distribution) shows remarkable differences between individuals, varying from 1.9 to 4.2 h, with an average value of 2.95 ± 0.92 h [9]. The t1/2 of the second phase of drug disappearance from the plasma (-phase, slow, elimination) varies even more, from 8.5 to 47 h [9,10,12,14–19]. Due to its high lipid solubility, 98% of methadone that has reached the central compartment is rapidly transferred to tissues, particularly liver, kidneys, lungs and, in small proportion, to the brain [20]; 1–2% remains in the blood compartment [9], 60–90% bound to plasma proteins, mostly the acid ␣1-globulins [21,22], and only in part (13.4–17.4%) to the ␥-globulins [23]. The extent of protein binding is of obvious importance for methadone activity. So, for example, the blood concentrations of acid ␣1 -glycoproteins, increase in stress conditions [23] and in heroin addicts. As a consequence, there is an increase of the amount of protein-bound methadone and a decrease of free and active methadone [24]. The size of the tissue compartment can be obtained by the apparent volume of distribution; in drug addicts this datum is not very consistent: 2.1–5.6 l kg−1 [9]; 4.1 l kg−1 [10] and 4.2–9.2 l kg−1 [20]. The large volume of methadone distribution indicates that there is a large sized tissue compartment, in dynamic equilibrium with a small central compartment. The result is that during maintenance treatment a short-lasting decrease of blood levels of methadone is usually not associated with clinically evident withdrawal symptoms [25]. During maintenance treatment, the plasma half-life of the -phase of elimination varies little in the same patient, even if the dose of methadone is increased or decreased, from 22 to 25 h, even though maintaining significant differences between individuals [26]. Some authors [13,27] are of the opinion that the induction of its own metabolism may be responsible for the reduction of methadone concentrations during maintenance treatment. According to Verebely [13] after thirty days of treatment at the daily dose of 40 or 80 mg, the plasma levels of methadone decrease by three to eight-times, while the excretion of the parent drug and of its metabolites increase from 22.2 to 61.9%. Accord- ing to Holmstrand [26], instead, the plasma concentrations of methadone decrease by 15–25% after 5–12 months of treatment with 60 or 80 mg/day by self-induction of its own metabolism, thus justifying the requests by some drug addicts to increase the daily dose in order to remain in treatment [28,29]. Individuals who relapse might have insufficient plasma levels of methadone, according to Nilsson [18]. This author examined the kinetics of methadone in a group of subjects under treatment and in a group of subjects who had withdrawn from treatment. In the patients of the latter group the volume of distribution and the t1/2 of the elimination -phase were shorter than in the patients of the former group (dropped-out patients: Vd: 3.09 ± 0.061 l kg−1 ; t1/2 : 24.5 ± 2.6 h; patients under treatment: Vd: 4.56 ± 1.001 l kg−1 ; t1/2 : 34.0 ± 7.0 h). Also the body clearance of methadone varies widely among individuals, ranging from 0.96 to 6.1 ml/min/kg [9]; thus, methadone body clearance, too, can contribute to the large differences in methadone kinetics among drug addicts. The role of chronic liver diseases in methadone metabolism is not clearly defined. Kreek [30,31] hypothesizes that the abnormal kinetics of methadone that are observed in some patients can be explained by the functional insufficiency of the hepatic microsomal system. The elimination of methadone and its metabolites occurs mainly through the kidneys: 15–60% during the first 24 h (20% as unmodified drug, and 13% as 2-ethylidene1,5-dimethyl-3,3-diphenylpyrrolidine; EDDP). Elimination in the faeces accounts for 20–40% [32]. According to Inturrisi [16] the overall elimination is 25% during the first 24 h, and 52% during the first 96 h (58% as methadone and 42% as metabolites). The rate of elimination of unmodified methadone is dependent on the extent of the pH-dependent tubular re-absorption in the kidney, and is increased by urine acidification. When urinary pH is less than 6, the amount of excreted methadone is three to eight-times greater than at pH higher than 6 [10,32–34]. Varying the pH also changes the half-life of plasma concentrations of methadone. Nilsson [32] evaluated the half-life of the -phase, the volume of distribution and body clearance after acidification (pH around 5.2) and alkalization (pH around 7.8) of the urines. The half time of the -phase was 19.5 ± 3.6 h with acidic urines, and 42.1 ± 8.8 with basic urines. Similar modifications have been found in the extent of the distribution volume, that goes from 3.51 ± 0.41 l kg−1 with acidic urines to 5.24 ± 0.83 l kg−1 with basic urines, and in body clearance, that goes from 134 ± 31 ml/min with acidic urines to 91.9 ± 9.1 ml/min with basic urines [32]. With acidic urines renal clearance triples, and the EDDP/methadone ratio decreases [34]. Urinary pH, therefore, has profound effects not only on methadone excretion, but also on the volume of distribution of the drug. By keeping the urinary pH constant, the inter-individual differences of methadone elimination and plasma concentrations are considerably reduced [15,32,34]. A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 3. Pharmacological interactions and cytochrome P450 (CYP) Changes in the metabolism and elimination of methadone are mainly caused by inhibition or induction of cytochrome P450, with a consequent increase or decrease of the amount of drug levels in blood and tissues. P450 and CYP are synonyms. A CYP enzyme is composed of a protein and of a haeme group (as the prosthetic group). This superfamily is divided into families and subfamilies of enzymes on the bases of their amino acid sequence. Each family has an identity of at least 40% in the amino acid sequence and is identified by an Arabic number. Each subfamily has an identity of at least 55% in the amino acid sequence and is indicated by an upper-case letter. The genes codifying a subfamily are identified by another Arabic number. For example, CYP3A4 means that the enzyme belongs to the family 3, to the subfamily A and is codified by the gene 4 [35]. Metabolic induction develops following the repeated administration of a drug, with the synthesis of new enzyme and with the increase of its activity. The result is an increase in the metabolism of the drug involved in interaction and a decrease in the quantity of drug available for pharmacological activity. In order for this to take place 1 or 2 weeks are usually needed. On the other hand, enzymatic inhibition develops quickly since it takes a short time for the drug to bind to the enzyme. Inhibition of activity of the enzyme decreases the metabolism of the drug, and therefore, increases its pharmacological activity [36]. Pharmacokinetic interaction can also occur when two or more drugs that are metabolic substrates of the same CYP are administered concurrently. In this case the drug that has the greatest affinity for that cytochrome can prevent in part the metabolism of the other drugs [37,38]. Most drugs are substrates of only five isoenzymes (CYP3A4, 1A2, 2C9, 2C19, 2D6); therefore, interactions can easily take place. The drugs that during absorption undergo a considerable first-pass effect or that have a low therapeutic index are the ones most often subject to significant interactions [39]. Many interactions are not clinically apparent because plasma concentrations with therapeutic doses are lower than those used to cause the interaction in vitro. 1 to 11-fold in the gut [42]. The major factor responsible for the variations of methadone bioavailability is the inter-individual difference in the expression of CYP3A4 [43]. This enzyme is involved also in the metabolism of other drugs: benzodiazepines, calcium antagonists, macrolide antibiotics, and anticonvulsants. Its activity is strongly inhibited by ketoconazole, by fluoxetine and by grapefruit juice (large amounts). Another enzyme, involved in methadone metabolism, is CYP2D6 [44]. It is mainly expressed in the liver, and is not inducible. This enzyme is subject to genetic polymorphism and many different CYP2D6 alleles have been identified [45]. The prevalence of the poor metabolism phenotype shows marked ethnic differences with a mean value of 7.4% (4–10%) of population in Europe and lower frequencies of 1% (0.6–1.5%) in Orientals [46,47]. The prevalence of rapid metabolizers is 1% of the German population, 7% of the Spanish and 2–5% of the Black population [47]. Also CYP1A2 is probably involved in methadone metabolism, but the available literature data are conflicting [47]. This isoenzyme is induced also by cigarette smoke, and is not subject to genetic polymorphism. Its activity can vary, according to the individual, Table 1 Drugs that are inducers or inhibitors of CYP3A4 and of CYP2D6 Inducers CYP3A4 Barbiturates Carbamazepine Dexamethasone Efavirenz Felbamate Hypericum (“St. John’s Worth”) Nelfinavir Nevirapine Oxcarbazepine Phenytoin Phosphophenytoin Rifampina Risperidone Ritonavir Topiramate Inhibitors Cimetidine Ciprofloxacin Clarithromycin Diltiazem Erythromycin Fluconazole Fluoxetine Fluvoxamine Grapefruit juice Josamycin Ketoconazolea Nefazodone Norfloxacin Norfluoxetine Paroxetine Protease Inhibitors Venlafaxine CYP2D6 (It can’t be induced.) Cimetidine Chlorimipramine Fluoxetine Fluvoxamine Haloperidol Levopromazine Moclobemide Norfluoxetine Paroxetine Protease Inhibitors Quinidinea Sertraline Thioridazine 4. Methadone metabolism Methadone is metabolised almost exclusively by the liver [10]. The main biotransformation of the two methadone enantiomers is the N-demethylation [40] by CYP3A4 [41]. CYP3A4 is found in the small intestine and in the liver; therefore, it affects both the intestinal and hepatic metabolism of methadone. This enzyme has no genetic polymorphism, it is inducible, and its activity varies greatly among individuals, from 1 to 30-fold in the liver, from 553 a Selective inducer or inhibitor. 554 Table 2 Drugs that are metabolic substrates of CYP3A4 and CYP2D6 Antidepressants Antipsychotics 3A4 Amitriptyline Imipramine Nefazodone Sertraline Venlafaxine Clozapine Haloperidol Risperidone 2D6 Desmethylcitalopram Fluoxetine Fluvoxamine Mianserine Nefazodone Norfluoxetine Paroxetine Sertraline Trazodone Venlafaxine Clozapine Haloperidol Perphenazine Risperidone Thioridazine a -Blockers Alprenolol Atenolol Metoprolola Propranolol Timolol Ca2+ -antagonists Benzodiazepines Antiarrhythmics Opioids Other drugs Amlodipine Diltiazem Nifedipine Nimodipine Verapamil Alprazolam Clonazepam Midazolama Triazolam Amiodarone Lidocaine Quinidine Tramadol Carbamazepine Clarithromycin Cortisol Cyclosporine Dexamethasone Erythromycina Ethinylestradiol Terfenadine Testosterone Theophylline Topiramate Troleandomycin Encainide Flecainide Propafenone Codeine Dextromethorphana Ethylmorphine Methadone Tramadol Amphetamines Chlorpromazine Sparteine Cinnarizine Flunarizine In vivo probes for studying induction and inhibition of cytochrome P450 enzymes in humans. A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 CYP A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 555 Table 3 Interactions between methadone, anticonvulsants and psychoactive drugs Drug Effect on methadone Amitriptyline [74] Carbamazepine [74] Diazepam/midazolam [74] Phenytoin/phosphophenytoin [75] Fluoxetine [49,50] Fluvoxaminea [49,50] Risperidone [76] Moclobemide [76] Plasma Plasma Plasma Plasma Plasma Plasma Plasma Plasma concentration concentration concentration concentration concentration concentration concentration concentration ↑, clearance ↓ ↓ ↑ ↓ (up to −50%) ↑ ↑ (+20 to 40%) ↓ ↑ Severity Onset Mechanism NR Moderate Moderate Moderate pWS Moderate Moderate Moderate Minor NR Delayed Delayed Delayed Rapid Rapid Delayed Rapid CYP2D6 substrate CYP3A4 induction CYP3A4 substrate CYP3A4 induction CYP3A4, CYP2D6 inhibition CYP3A4, CYP2D6 inhibition CYP3A4 induction CYP2D6, CYP1A2 inhibition NR: not reported; ↑: increase; ↓: decrease; pWS: probable Withdrawal Symptoms. Moderate severity: it can worsen the patient’s condition and/or it requires a change in the therapy; minor severity: clinical effects are limited; it can increase the frequency and/or the seriousness of side effects, but no medical intervention is necessary. Delayed onset: appearance of clinical effects after more than 24 h from the administration of the drug; rapid onset: appearance of clinical effects within 24 h from the administration of the drug. a The discontinuance of treatment has been associated with withdrawal symptoms. from 1 to 40-fold. The expression of these three enzymes in the liver is quite different; CYP3A4, CYP2D6, CYP1A2 represent, respectively, 30, 4 and 13% of all CYP enzymes [42]. Tables 1 and 2 list the inducer, inhibitor and metabolic substrate drugs of CYP3A4 and CYP2D6. 5. Interactions of methadone Methadone maintenance treatment must not be interrupted too early. In fact, its aim is to retain drug addicts on treatment for months or years [2]. During these long periods, treatments with other drugs may become necessary in consideration of the high comorbidity of drug addicts [48], and there may be the risk of drug–drug interactions. The classes of drugs that could be used during methadone maintenance treatment and that could produce drug–drug interactions— of the kinetic type—with methadone, are benzodiazepines, antidepressants, anticonvulsants (Table 3), macrolide antibiotics and anti-fungals (Table 4). These drugs are inhibitors, inducers or substrates of CYP3A4 or CYP2D6. Particular mention is made of the interaction with the antidepressant fluvoxamine, which inhibits both CYP3A4 and 2D6, and which attains in vivo, at therapeutic doses [48], plasma concentrations that correspond to those that are inhibitory in vitro [49,50]. Another clinically important interaction may occur with rifampicin: in order to avoid withdrawal symptoms an increase of up to double the methadone dose [51,52] is required within a few days after the start of rifampicin treatment. Maintenance treatment with methadone is best choice in HIV positive heroin addicts, therefore, the interactions that can take place most often and that are clinically most important are those between methadone and antiretroviral drugs [53–59] (Table 5). In HIV therapy, three classes of antiretroviral drugs are currently used: (1) the nucleotide or nucleoside analogues, that are inhibitors of the reverse transcriptase (zidovudine, didanosine, zalcitabine, lamivudine, abacavir); (2) the non-nucleoside inhibitors of the reverse transcriptase (nevirapine, delavirdine, efavirenz); (3) the protease inhibitors (saquinavir, ritonavir, indinavir, nelfinavir, amprenavir) [60–62]. The most frequently used associations include two nucleoside analogues and a protease inhibitor, or two nucleoside analogues and a non nucleoside inhibitor of the reverse transcriptase [60,61]. Antiretrovirals are metabolic inducers of CYP3A4 and this entails an Table 4 Interactions between methadone and drugs or substances of possible use in heroin addicts Drug Effect on methadone Severity Onset Mechanism Ketoconazole [77] Dexamethasone [78] Disulfiram [54] Fluconazole [77,79] Erythromycin [80] Rifampin [51,52,81] Rifabutine [52] Grapefruit juicea [82] Plasma concentration ↑ Plasma concentration ↓ Plasma concentration ↓ Bioavalability ↑ (AUC: +35%; clearance: −24%) Plasma concentration ↑ Plasma concentration ↓ (−30%, −65%) Plasma concentration ↓ Plasma concentration ↑ Minor NE NE Minor Moderate Moderate; pWS Moderate Moderate NR NR NR NR Delayed Delayed (6–8 h) Delayed Rapid Strong CYP3A4 inhibition CYP3A4 induction NR CYP3A4 inhibition CYP3A4 inhibition Strong CYP3A4 induction CYP3A4 induction CYP3A4 inhibition NE: no effect; NR: not reported; ↑: increase; ↓: decrease; pWS: probable Withdrawal Symptoms. Moderate severity: it can worsen the patient’s condition and/or it requires a change in the therapy; minor severity: clinical effects are limited; it can increase the frequency and/or the seriousness of side effects, but no medical intervention is necessary. Delayed onset: appearance of clinical effects after more than 24 h from the administration of the drug; rapid onset: appearance of clinical effects within 24 h from the administration of the drug. a The increase of methadone concentration is clinically significant only after the ingestion of large amounts of grapefruit juice, several times a day and for several days. 556 A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 Table 5 Interactions between methadone and antiretroviral drugs Drug Effect on methadone Abacavir [83] Amprenavir [60,83] Didanosine [84,85] Efavirenz [67–69] Indinavir [60] Nelfinavir [61] Nevirapine [66,86] Ritonavir [87] Stavudine [84] Zidovudine [88] Plasma Plasma Plasma Plasma Plasma Plasma Plasma Plasma Plasma Plasma concentration concentration concentration concentration concentration concentration concentration concentration concentration concentration ↓ ↓ ↓ ↓ ↑ ↓ ↓ ↓ ↓ ↑ (Cmax : −34%) (−60%) (29–47%) (−46%) (−40 to 50%) Severity Onset Mechanism Minor Minor Minor Moderate; NE Moderate; Moderate; Moderate; Minor Moderate NR Delayed Rapid Delayed (8–10 days) NR Delayed Delayed (4–8 days) Delayed Rapid Delayed M clearance ↑ (+22%) NR Bioavailability ↓ (−41 to 59%) M metabolism ↑ (CYP3A4 induction) Bioavailability ↑ M metabolism ↑ (CYP3A4 induction) M metabolism ↑ (CYP3A4 induction) M metabolism ↑ (CYP3A4 induction) NR M bioavailability ↑ pWSa pWSa pWSa pWSa M: methadone; NE: no effect; NR: not reported; ↑: increase; ↓: decrease; pWS: probable Withdrawal Symptoms. Moderate severity: it can worsen the patient’s condition and/or it requires a change in the therapy; minor severity: clinical effects are limited; it can increase the frequency and/or the seriousness of side effects, but no medical intervention is necessary. Delayed onset: appearance of clinical effects after more than 24 h from the administration of the drug; rapid onset: appearance of clinical effects within 24 h from the administration of the drug. a During the methadone treatment it is recommended to check withdrawal symptoms and to increase the dosage on the basis of the intensity of withdrawal symptoms. Table 6 Interactions between methadone and combinations of antiretroviral drugsa Drugs Effect on methadone Ritonavir + lopinavir [89–91] Ritonavir + saquinavir [92] Ritonavir + nelfinavir [56] Ritonavir + nelfinavir + nevirapine [64] Nevirapine + efavirenz [64] Abacavir + amprenavir [83] Plasma Plasma Plasma Plasma Plasma Plasma concentration concentration concentration concentration concentration concentration Severity ↓ (AUC: −30%; Cmax : −44%) ↓ (−28%) ↓ ↓ ↓ (AUC: −54%; Cmax : −42%) ↓ Minor Moderate; pWS Moderate; pWS Minor Minor Minor ↓: decrease; pWS: probable Withdrawal Symptoms. Moderate severity: it can worsen the patient’s condition and/or it requires a change in the therapy; minor severity: clinical effects are limited; it can increase the frequency and/or the seriousness of side effects, but no medical intervention is necessary. a For all these interactions the onset is delayed and the probable mechanism is an increased metabolism of methadone; during the methadone treatment it is recommended to monitor withdrawal symptoms and to increase the dosage on the basis of the intensity of withdrawal symptoms. increase in enzymatic activity, a decrease of the amount of methadone available, and the possible precipitation of opioid withdrawal symptoms [63,64] (Table 6). Because of the extreme variability of methadone metabolism in the same subject and among different subjects, the relationship between dose–blood level-effect is not established [65] and it is impossible to suggest guidelines of management in order to prevent the onset of the opioid withdrawal syndrome during polytherapy. The degree of the increase of methadone dosage after the occurrence of withdrawal symptoms in the course of anti-HIV treatment has been extremely different [55,66,67] even when the antiretroviral drug used was the same (Table 7). Also the time of onset and the severity of the withdrawal syndrome were different, in the above-quoted studies, perhaps because the patients had been treated for different periods of time and with extremely different daily doses of methadone. The interaction between efavirenz and methadone, for example, determined a decrease in the plasma concentrations of methadone that varied between 22 and 80%, with signs and symptoms of morphine withdrawal that appeared from 4 to 28 days after the beginning of the treatment with the antiretroviral drug [67–69]. Table 7 Interactions between methadone and antiretroviral drugs, and measures taken to abolish withdrawal symptomsa Drug Effect on methadone Days after the beginning of treatment Measures taken to treat withdrawal symptoms Efavirenz [67] Efavirenz [56] Bioavailability ↓ (on average: −60%) Plasma concentration ↓ (on average: −40% in patients treated with M 100 mg/day for 1 year) Plasma concentration ↓ (on average: −65%) Plasma concentration ↓ (in patients treated with M 90 mg/day for 2 years) 8–10 28 M dosage ↑ (+66–133%) M dosage ↑ (up to 180 mg/day) 8–10 7 M dosage ↑ (+65–133%) M dosage ↑ (up to 130 mg/day) Efavirenz [69] Ritonavir + saquinavir [92] M: methadone;↑: increase; ↓: decrease. a It is advised to check frequently withdrawal symptoms. A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 557 In conclusion, it can only be recommended a careful observation of the patient and, if necessary, an adjustment of the daily dose of methadone on the basis of the clinical signs. It is well-known that during methadone maintenance treatment many subjects increase their alcohol consumption [70]; when chronically abused, alcohol induces the activity of CYP3A4 [71]. In such circumstances the plasma concentrations of drugs that are metabolised by this enzymes (particularly methadone), decrease [72]. Other opioid drugs, such as codeine and tramadol, are metabolised by the same enzymes, and thus are subjected to the same interactions as methadone. In particular, buprenorphine is metabolised by CYP3A4 [73] and should undergo exactly the same interactions as methadone. Data on this subject are anectodal because the use of buprenorphine on a large scale in the treatment of heroin addicts started in France and Italy only a few years ago. However, there is a clear evidence that the interactions of buprenorphine with antiretroviral drugs are similar to those of methadone [50]: its plasma concentrations may decrease when it is administered together with protease inhibitors or other enzyme inducers. As a rule, in order to prevent, identify and quickly manage pharmacological interactions, the physicians should: 6. Conclusions References The possibility that clinically important interactions occur when methadone is taken concomitantly with other drugs is substantial. Fortunately, most of such pharmacokinetic interactions are not life-threatening; however, they can have important consequences: precipitation of withdrawal symptoms, relapse in the use of heroin in an attempt to relieve them, thus leaving the maintenance treatment. Physicians must, therefore, carefully follow these patients in order to avoid, or at least to notice and treat in time, such plights. Since the pharmacokinetics of methadone is extremely variable from one patient to another, the relationships between dose, plasma levels and effects are not clearly defined, and an optimum range of therapeutic concentrations has not yet been identified for maintenance treatment. The same dose, even though normalized to weight, produces a bioavailability of methadone that is completely different from subject to subject. Thus, in the course of long-term maintenance treatments, the daily dose must be personalised; using doses of methadone in an arbitrary manner, within a limited range, is clinically incorrect. In choosing the dose, the simultaneous use of other drugs must be taken into careful consideration. The effectiveness of maintenance treatment must be evaluated over time. To do this, since it isn’t always possible to measure the blood levels of methadone (that, in addition, are not always sufficient to predict the effects of the drug if it not associated with other pharmacokinetic parameters) all that can be done is to check the urine for drugs of abuse and to carefully consider drug addicts’ self-reports. Only on the basis of these elements it will be possible to adjust the dosage of methadone to the real needs of the patient under treatment. [1] Dole VP, Nyswander ME. Heroin addiction—a metabolic disease. Arch Intern Med 1967;120:19–24. [2] Brettle RP. HIV and harm reduction for injection drug users. AIDS 1991;5:125–36. [3] Leshner AI. Drug abuse and addiction treatment research: the next generation. Arch Gen Psychiatry 1997;54:691–4. [4] Ward J, Bell J, Mattick RP, Hall W. Methadone maintenance therapy for opioid dependence. CNS Drugs 1996;6:440–9. [5] Scott CC, Robbins EB, Chen KK. Pharmacological comparison of the optical isomers of methadone. J Pharmacol Exp Ther 1948;92:282–6. [6] Olsen GD, Wendel HA, Livermore JD, Leger RM, Lynn RK, Gerber N. Clinical effect and pharmacokinetics of racemic methadone and its optical isomers. Clin Pharmacol Ther 1977;21:147–57. [7] Kristensen K, Christensen CB, Christrup LL. The mu 1, mu 2, delta, kappa opioid receptor binding profiles of methadone stereoisomers and morphine. Life Sci 1995;56:PL45–50. [8] Inturrisi CE, Verebely K. Disposition of methadone in man after a single oral dose. Clin Pharmacol Ther 1972;13:923–30. [9] Meresaar U, Nilsson MI, Holmstrand J, Anggard E. Single dose pharmacokinetics and bioavailability of methadone in man studied with a stable isotope method. Eur J Clin Pharmacol 1981;20:473–8. [10] Nilsson MI, Meresaar U, Anggard E. Clinical pharmacokinetics of methadone. Acta Anaesthesiol Scand 1982;74:66–9. [11] de Vos JW, Geerlings PJ, Van der Brink W, Ufkes JG, Van Wilgenburg H. Pharmacokinetics of methadone and its primary metabolite in 20 opiate addicts. Eur J Clin Pharmacol 1995;48:361–6. [12] Wolff K, Sanderson M, Hay AW, Raistrick D. Methadone concentrations in plasma and their relationship to drug dosage. Clin Chem 1991;37:205–9. [13] Verebely K, Volavka J, Mule S, Resnick R. Methadone in man: pharmacokinetic and excretion studies in acute and chronic treatment. Clin Pharmacol Ther 1975;18:180–90. [14] Anggard E, Nilsson MI, Holmstrand J, Gunne LM. Pharmacokinetics of methadone during maintenance: pulse labeling with deuterated methadone in the steady-state. Eur J Clin Pharmacol 1979;16:53–75. [15] Inturrisi CE, Verebely K. The levels of methadone in the plasma in methadone maintenance. Clin Pharmacol Ther 1972;13:633–7. (1) record all drugs and any abuse substances, including alcohol, that the patient under methadone maintenance is taking; this list should always be consulted before prescribing a new drug; (2) know the pharmacodynamics and the pharmacokinetics of the drugs that he prescribes, bearing in mind the potential mechanisms of pharmacological interactions; (3) be particularly watchful towards patients with illnesses, such as kidney or liver insufficiency, that could modify drug kinetics and dynamics; (4) look for any possible drug interaction whenever the patient complains of withdrawal symptoms or excessive sedation, or unusual symptoms with respect to the dosage of the drugs he is taking, or to other concurrent pathologies; (5) watch for any interactions, not yet mentioned in the literature, when administering drugs recently introduced on the market. 558 A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 [16] Inturrisi CE, Colburn WA, Kaiko RF, Houde RW, Foley KM. Pharmacokinetics and pharmacodynamics of methadone in patients with chronic pain. Clin Pharmacol Ther 1987;41:392–401. [17] Loimer N, Schmid R, Grunberger J, Jagsch R, Linzmayer L, Presslich O. Psychophysiological reactions in methadone maintenance patients do not correlate with methadone plasma levels. Psychopharmacology (Berl) 1991;103:538–40. [18] Nilsson MI, Gronbladh L, Widerlov E, Anggard E. Pharmacokinetics of methadone in methadone maintenance treatment: characterization of therapeutic failures. Eur J Clin Pharmacol 1983;25:497–501. [19] Wolff K, Hay AW, Raistrick D, Calvert R. Steady-state pharmacokinetics of methadone in opioid addicts. Eur J Clin Pharmacol 1993;44:189–94. [20] Dole VP, Kreek MJ. Methadone plasma level: sustained by a reservoir of drug in tissue. Proc Natl Acad Sci USA 1973;70:10. [21] Eap CB, Cuendet C, Baumann P. Binding of d-methadone, lmethadone, and dl-methadone to proteins in plasma of healthy volunteers: role of the variants of alpha l-acid glycoprotein. Clin Pharmacol Ther 1990;47:338–46. [22] Garrido MJ, Aguirre C, Troconiz IF, Marot M, Valle M, Zamacona MK, et al. Alpha-1 acid glycoprotein (AAG) and serum protein binding of methadone in heroin addicts with abstinence syndrome. J Clin Pharmacol Ther 2000;38:35–40. [23] Olsen GD. Methadone binding to human plasma proteins. Clin Pharmacol Ther 1973;14:338–43. [24] Garrido MJ, Jimenez R, Gomez E, Calvo R. Influence of plasma– protein binding on analgesic effect of methadone in rats with spontaneous withdrawal. J Pharm Pharmacol 1996;48:281–4. [25] Plummer JL, Gourlay GK, Cherry DA, Cousins MJ. Estimation of methadone clearance: application in the management of cancer pain. Pain 1988;33:313–22. [26] Holmstrand J, Anggard E, Gunne LM. Methadone maintenance: plasma levels and therapeutic outcome. Clin Pharmacol Ther 1978;23:175–80. [27] Anggard E, Gunne LM, Holmstrand J, McMahon RE, Sandberg CG, Sullivan HR. Disposition of methadone in methadone maintenance. Clin Pharmacol Ther 1975;17:258–66. [28] Horns WH, Rado M, Goldstein A. Plasma levels and symptom complaints in patients maintained on daily dosage of methadone hydrochloride. Clin Pharmacol Ther 1975;17:636–49. [29] Nilsson MI, Anggard E, Holmstrand J, Gunne LM. Pharmacokinetics of methadone during maintenance treatment: adaptive changes during the induction phase. Eur J Clin Pharmacol 1982;22:343–9. [30] Kreek MJ. Plasma and urine levels of methadone. Comparison following four medication forms used in chronic maintenance treatment. NY State J Med 1973;73:2773–7. [31] Kreek MJ. Methadone in treatment: physiological and pharmacological issues. Handbook on drug abuse. Washington DC: US Government Printing Office; 1979. p. 57–86. [32] Nilsson MI, Widerlov E, Meresaar U, Anggard E. Effect of urinary pH on the disposition of methadone in man. Eur J Clin Pharmacol 1982;22:337–42. [33] Baselt RC, Casarett LJ. Urinary excrection of methadone in man. Clin Pharmacol Ther 1972;13:64–70. [34] Bellward GD, Warren PM, Howald W, Axelson JE, Abbott FS. Methadone maintenance: effect of urinary pH on renal clearance in chronic high and low doses. Clin Pharmacol Ther 1977;22:92–9. [35] Nebert DW, Nelson DR, Coon MJ, Estabrook RW, Feyereisen R, Fuji-Kuriyama Y, et al. The P450 superfamily: update on new sequences, gene mapping and recommended nomenclature. DNA Cell Biol 1991;10:1–14. [36] Lin JH, Lu AY. Inhibition and induction of cytochrome P450 and the clinical implications. Clin Pharmacokinet 1998;35:361–90. [37] Shou M, Grogan J, Mancewicz JA, Krausz KW, Gonzalez FJ, Gelboin HV, et al. Activaction of CYP 3A4: evidence for the simultaneous binding of two substrates in a cytochrome P450 active site. Biochemistry 1994;33:6450–5. [38] Korzekwa KR, Krishnamachary N, Shou M, Ogai A, Parise RA, Rettie AE, et al. Evaluation of atypical cytochrome P450 kinetics with two-substrate models: evidence that multiple substrates can simultaneously bind to cytochrome P450 active sites. Biochemistry 1998;37:4137–47. [39] Anonymous. FDA position on product selection for “narrow therapeutic index” drugs. Am J Health Syst Pharm 1997;54:1630–2. [40] Moody DE, Alburges ME, Parker RJ, Collins JM, Strong JM. The involvement of cytochrome P450 3A4 in the N-demethylation of l-alpha-acetylmethadol (LAAM), norLAAM, and methadone. Drug Metab Dispos 1997;25:1347–53. [41] Beckett AH, Taylor JF, Casy AF, Hassan MM. The biotransformation of methadone in man: synthesis and identification of a major metabolite. J Pharm Pharmacol 1968;20:754–62. [42] Ketter TA, Flockhart DA, Post RM, Denicoff K, Pazzaglia PJ, Marangell LB, et al. The emerging role of cytochrome P450 3A in psychopharmacology. J Clin Psychopharmacol 1995;15:387–98. [43] Garrido MJ, Troconiz IF. Methadone: a review of its pharmacokinetic/pharmacodynamic properties. J Pharmacol Toxicol Methods 1999;42:61–6. [44] Wu D, Otton SV, Sproule BA, Busto U, Inaba T, Kalow W, et al. Inhibition of human cytochrome P450 2D6 (CYP2D6) by methadone. Br J Clin Pharmacol 1993;35:30–4. [45] Begre S, von Bardeleben U, Ladewig D, Jaquet-Rochat S, CosendaiSavary L, Golay KP, et al. Paroxetine increases steady-state concentrations of (R)-methadone in CYP2D6 extensive but not poor metabolizers. J Clin Psychopharmacol 2002;22:211–5. [46] Eap CB, Broly F, Mino A, Hammig R, Deglon JJ, Uehlinger C, et al. Cytochrome P450 2D6 genotype and methadone steady-state concentrations. J Clin Psycopharmacol 2001;21:229–34. [47] Nakamura K, Goto F, Ray WA, Mc Allister CB, Jacqz E, Wilkinson GR, et al. Indomethacin differences in genetic polymorphism of debrisoquine and mephenytoin hydroxylation between Japanese and Caucasian population. Clin Pharmacol Ther 1985;38:402–8. [48] O’Connor PG, Fiellin DA. Pharmacologic treatment of heroindependent patients. Ann Intern Med 2000;133:40–54. [49] Eap CB, Bertshy G, Powell K, Baumann P. Fluvoxamine and fluoxetine do not interact in the same way with the metabolism of the enantiomers of methadone. J Clin Psychopharmacol 1997;17:113–7. [50] Iribarne C, Picart D, Dreano Y, Berthou F. In vitro interactions between fluoxetine or fluvoxamine and methadone or buprenorphine. Fundam Clin Pharmacol 1998;12:194–9. [51] Kreek MJ, Garfield JW, Gutjahr CL, Giusti LM. Rifampin-induced methadone withdrawal. N Engl J Med 1976;294:1104–6. [52] Baciewicz AM, Self TH. Rifampin drug interactions. Arch Intern Med 1984;144:1667–71. [53] Furlan V, Taburet AM. Drug interactions with antiretroviral agents. Therapie 2001;56:267–71. [54] Vastag B. Health agencies update. HIV and heroin interactions. J Am Med Assoc 2001;286:295. [55] Gourevitch MN. Interactions between HIV-related medications and methadone: an overview. Updated March 2001. Mt Sinai J Med 2001;68:227–8. [56] Khalsa J, Genser S, Vocci F, Francis H, Bean P. The challenging interactions between antiretroviral agents and addiction drugs. Am Clin Lab 2002;21:10–3. [57] McCance-Katz EF, Gourevitch MN, Arnsten J, Sarlo J, Rainey P, Jatlow P. Modified directly observed therapy (MDOT) for injection drug users with HIV disease. Am J Addict 2002;11:271–8. [58] Perez Pons JC, Jornet Montana S, Bonet Esteve A. Pharmacokinetic interactions between methadone and antiretroviral medication in HIV positive patients. Med Clin (Barc) 2002;119:224–9. [59] Rainey PM. HIV drug interactions: the good, the bad and the other. Ther Drug Monit 2002;24:26–31. [60] Barry M, Gibbons S, Back D, Mulcahy F. Protease inhibitors in patients with HIV disease. Clinically important pharmacokinetic considerations. Clin Pharmacokinet 1997;32:194–209. A. Ferrari et al. / Pharmacological Research 50 (2004) 551–559 [61] Beauverie P, Taburet AM, Dessalles MC, Furlan V, Touzeau D. Therapeutic drug monitoring of methadone in HIV-infected patients receiving protease inhibitors. AIDS 1998;12:2510–1. [62] Iribarne C, Berthou F, Carlhant D, Dreano Y, Picart D, Lohezic F, et al. Inhibition of methadone and buprenorphine N-dealkylations by three HIV-1 protease inhibitors. Drug Metab Dispos 1998;26:257–60. [63] Borg L, Kreek MJ. Clinical problems associated with interactions between methadone pharmacotherapy and medications used in the treatment of HIV positive and AIDS patients. Curr Opin Psychiatry 1995;8:199–202. [64] Akerele EO, Levin F, Nunes E, Brady R, Kleber H. Effects of HIV triple therapy on methadone levels. Am J Addict 2002;11:308–14. [65] Horns WH, Rado M, Goldstein A. Plasma levels and symptoms complaints in patients maintained on daily dosage of methadone hydrochloride. Clin Pharmacol Ther 1975;17:636–49. [66] Clarke SM, Mulcahy FM, Tjia J, Reynolds HE, Gibbons SE, Barry MG, et al. Pharmacokinetic interactions of nevirapine and methadone and guidelines for use of nevirapine to treat injection drug users. Clin Infect Dis 2001;33:1595–7. [67] Boffito M, Rossati A, Reynolds HE, Hoggard PG, Back DJ, Di Perri G. Undefined duration of opiate withdrawal induced by efavirenz in drug users with HIV infection and undergoing chronic methadone treatment. AIDS Res Hum Retroviruses 2002;18:341–2. [68] Clarke S, Mulcahy F, Bergin C, Reynolds H, Boyle N, Barry M, et al. Absence of opioid withdrawal symptoms in patients receiving methadone and the protease inhibitor lopinavir–ritonavir. Clin Infect Dis 2002;34:1143–5. [69] Calvo R, Lukas JC, Rodriguez M, Carlos MA, Suarez E. Pharmacokinetics of methadone in HIV-positive patients receiving the non-nucleoside reverse transcriptase efavirenz. Br J Clin Pharmacol 2002;53:212–4. [70] Ottomanelli G. Methadone patients and alcohol abuse. J Subst Abuse Treat 1999;16:113–21. [71] Kreek MJ. Metabolic interactions between opioids and alcohol. Ann NY Acad Sci 1981;362:36–49. [72] Klotz U, Ammon E. Clinical and toxicological consequences of the inductive potential of ethanol. Eur J Clin Pharmacol 1998;54:7–12. [73] Ibrahim RB, Wilson JG, Thorsby ME, Edwards DJ. Effect of buprenorphine on CYP 3A activity in rat and human microsomes. Life Sci 2000;66:1293–8. [74] Preston KL, Griffiths RR, Stitzer ML, Bigelow GE, Liebson IA. Diazepam and methadone interaction in methadone maintenance. Clin Pharmacol Ther 1984;36:534–41. [75] Tong TG, Pond SM, Kreek MJ, Jaffery NF, Benowitz NL. Phenytoininduced methadone withdrawal. Ann Intern Med 1981;94:349–51. [76] Markowitz JS, Wells BG, Carson WH. Interactions between antipsychotic and antihypertensive drugs. Ann Pharmacother 1995;29:603– 9. 559 [77] Katz HI. Drug interactions of the newer oral antifungal agents. Br J Dermatol 1999;141:26–32. [78] Villikka K, Kivisto KT, Neuvonen PJ. The effect of dexamethazone on the pharmacokinetics of triazolam. Pharmacol Toxicol 1998;83:135–8. [79] Tarumi Y, Pereira J, Watanabe S. Methadone and fluconazole: respiratory depression by drug interaction. J Pain Symptom Manage 2002;23:148–53. [80] Gurevitz SL. Erythromycin: drug interactions. J Dent Hyg 1997;71:159–61. [81] Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivisto KT. Pharmacokinetic interactions with rifampicin: clinical relevance. Clin Pharmacokinet 2003;42:819–50. [82] Fuhr U. Drug interactions with grapefruit juice. Drug Saf 1998;18:251–72. [83] Bart PA, Rizzardi PG, Gallant S, Golay KP, Baumann P, Pantaleo G, et al. Methadone blood concentrations are decreased by the administration of abacavir plus amprenavir. Ther Drug Monit 2001;23:553–5. [84] Rainey PM, Friedland G, McCance-Katz EF. Interaction of methadone with didanosine and stavudine. J Acquir Immune Defic Syndr 2000;24:241–8. [85] Gourevitch MN, Friedland GH. Interactions between methadone and medications used to treat HIV infection: a review. Mt Sinai J Med 2000;67:429–36. [86] Altice FL, Friedland GH, Cooney EL. Nevirapine induced opiate withdrawal among injection drug users with HIV infection receiving methadone. AIDS 1999;13:957–62. [87] Geletko SM, Erickson AD. Decreased methadone effect after ritonavir initiation. Pharmacotherapy 2000;20:93–4. [88] McCance-Katz EF, Rainey PM, Friedland G, Kosten TR, Jatlow P. Effect of opioid dependence pharmacotherapies on zidovudine disposition. Am J Addict 2001;10:296–307. [89] Clarke SM, Mulcahy F, Bergin C, Reynolds H, Boyle N, Barry M, et al. Absence of opioid withdrawal symptoms in patients receiving methadone and the protease inhibitor lopinavir–ritonavir. Clin Infect Dis 2002;34:1143–5. [90] McCance-Katz EF, Rainey PM, Friedland G, Jatlow P. The protease inhibitor lopinavir-ritonavir may produce opiate withdrawal in methadone maintained patients. Clin Infect Dis 2003;37:476– 82. [91] Stevens RC, Rapaport S, Maroldo-Connelly L, Patterson JB, Bertz R. Lack of methadone dose alterations or withdrawal symptoms during therapy with lopinavir/ritonavir. J Acquir Immune Defic Syndr 2003;33:650–1. [92] Gerber JG, Rosenkranz S, Segal Y, Aberg J, D’Amico R, Mildvan D, et al. Effect of ritonavir/saquinavir on stereoselective pharmacokinetics of methadone: results of AIDS Clinical Trials Group (ACTG) 401. J Acquir Immune Defic Syndr 2001;27:153–60.