You are on page 1of 6

CE: M.S.

; JAM-D-17-00166; Total nos of Pages: 6;


JAM-D-17-00166

ORIGINAL RESEARCH

The Use of Serum Methadone/Metabolite Ratios to


Monitor Changing Perinatal Pharmacokinetics
John J. McCarthy, MD, Ernest J. Vasti, MD, Martin H. Leamon, MD,
Joseph Graas, PhD, Coburn Ward, PhD, and Catherine Fassbender, PhD

Objectives: Pregnancy profoundly alters drug metabolism, acceler-


ating clearance and confounding medication management, primarily
T here has been a dramatic increase over the past decade in
the United States in the use of opioids during pregnancy
(whether related to opioid use disorder or pain management),
through induction of CYP450 enzymes. Methadone is a CYP450 and a concurrent increase in newborns developing neonatal
substrate with altered pharmacokinetics during pregnancy. We report abstinence syndrome (NAS) (Corr and Hollenbeak, 2017).
on the use of serum methadone/metabolite ratios (MMRs) to monitor This makes improvements in management of dependence
changes in methadone metabolism through the perinatal period and during pregnancy an urgent priority. A recent Cochrane report
to objectively guide methadone dosing. Previous research found identified methadone and buprenorphine as effective medi-
average MMRs in nonpregnant populations of between 11.3 and cations for treatment of opioid use disorder, but only under
12.7. conditions of adequate dosing (Mattick et al., 2014). However,
Methods: Serum methadone and its major metabolite 2-ethylidene- at fixed doses of methadone, as the pregnancy progresses,
1,5-dimethyl-3,3-diphenylpyrrolidine concentrations were analyzed most pregnant women will experience recurrent withdrawal
in 67 samples from 23 pregnant patients treated for opioid use symptoms. In addition to threatening a woman’s sobriety,
disorder, and their calculated ratio was used to document changes withdrawal may result in increases in maternal stress hormone
in methadone clearance across trimesters and postpartum. Lower activity (catecholamines and corticosteroids) that have poten-
ratios indicate increased clearance. tial adverse consequences both for neonatal and long-term
Results: The average MMR during pregnancy was 6.1. Ratios health (McCarthy et al., 2017).
declined significantly from trimester 1 to trimester 3 (P ¼ 0.007), Pharmacogenetics, the study of individual variations in
and then rose significantly from trimester 3 to postpartum drug response based on a subject’s genetic profile, has impor-
(P ¼ 0.001). The per cent of ratios that were 4 or less, indicating tant implications for safety and efficacy of medication usage
ultrarapid metabolism, increased from 8% to 30% to 38% across (Weinshilboum, 2003). Methadone is primarily metabolized
trimesters, and decreased to 5% postpartum. Forty-four per cent of
by cytochrome P450 (CYP) enzymes 2B6, 3A4, and by lesser
and variable contributions from 2D6, 2C19, 2C9, and 1A2,
individual patients had at least 1 prepartum ratio of 4 or less.
with the parent molecule being demethylated into inactive 2-
Conclusions: This study documents significant metabolic changes
ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP)
occurring perinatally, which indicate the need for both changes in
(Eap et al., 2002; Kharasch et al., 2009). CYP genetic
methadone dose and dose frequency to maintain maternal/fetal polymorphism contributes to a 17-fold variation in methadone
stability, and also dose reductions as hypermetabolism reverses serum concentration for a given dosage (Eap et al., 2002). This
postpartum. MMRs provide an objective tool to more efficiently metabolic system’s ancestral gene is more than 2 billion years
improve the safety and efficacy of methadone dosing perinatally. old, but its diversity accelerated 800 million years ago as a
Key Words: CYP450, metabolic ratio, methadone/metabolite, result of animal/plant competitive evolution: plants producing
pharmacokinetics, pregnancy protective toxins and animals, who eat the plants, producing
enzymes to metabolize the toxins (Nebert et al., 1989). The
(J Addict Med 2018;xx: xxx–xxx) expansion of the P450 system also provided an evolutionary
advantage in protecting the animal offspring (and ultimately
the human fetus) from toxins and xenobiotics. However, this
From the Department of Psychiatry, University of California, Davis, CA also creates significant challenges to safe and efficacious
(JJMC, MHL, CF); Department of Family Medicine, University of medication management perinatally for many medications,
California, Davis, CA (EJV); San Diego Reference Laboratory, San
Diego, CA (JG); and University of the Pacific, Stockton, CA (CW).
including methadone.
Received for publication October 14, 2017; accepted February 8, 2018. Superimposed on this background of genetic variability
The authors report no conflicts of interest. are metabolic changes during the perinatal period which
Send correspondence to John J. McCarthy, MD, PO Box 561, Tahoma, CA further alter medication pharmacokinetics through changes
96142. E-mail: drjackmac9@gmail.com.
Copyright ß 2018 American Society of Addiction Medicine
in blood volume, absorption, protein binding, metabolism,
ISSN: 1932-0620/16/0000-0001 and elimination (Jeong, 2010). However, these changes are
DOI: 10.1097/ADM.0000000000000398 less relevant than the most important change related to

J Addict Med  Volume 00, Number 00, Month/Month 2018 1

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.
CE: M.S.; JAM-D-17-00166; Total nos of Pages: 6;
JAM-D-17-00166

McCarthy et al. J Addict Med  Volume 00, Number 00, Month/Month 2018

medication use, which is the pregnancy-related activation of We added EDDP serum levels assays to our clinic’s
genes coding for the CYP enzymes (Weinshilboum, 2003). routine methadone serum protocol to assess the relevance and
Changing levels of pregnancy hormones induce gene expres- utility of metabolic ratios in actual patient care. This paper
sion and regulate P450 RNA, which drives the altered phar- will report the use of sequential MMRs during pregnancy and
macokinetics (Isoherranen and Thummel, 2013). There is the postpartum period to monitor pharmacokinetic changes as
evidence of induction of both CYP2D6 (Wadelius et al., an additional aid to safe and effective individualized dosing.
1997) and CYP3A4 (Isoherranen and Thummel, 2013) by
progesterone in the luteal phase of the menstrual cycle, and, if METHODS
this phase ends in pregnancy, the CYP system has already Starting in January 2015, serum methadone and EDDP
been primed for the profound changes in drug metabolism and levels and ratio data were collected from pregnant patients
transport that occur during pregnancy. Pregnancy specifically enrolled in a specialized pregnancy program within a metha-
induces CYP3A4 (Tracy et al., 2005), 2D6 (Wadelius et al., done maintenance program. Routine care involved weekly
1997), and 2B6 (Dickmann and Isoherranen, 2013). Changes meetings with a pregnancy counselor, attendance at weekly
in pregnancy can shorten the effective half-life of methadone physician-guided pregnancy groups, and monthly individual
from its more usual 24-hour range to 12 hours (Bogen et al., meetings with the clinic physician. Serum level and ratio data
2013), and at times to as short as 4 to 6 hours (McCarthy et al., were routinely collected as clinically determined every 4 to
2015). 6 weeks during the pregnancy and within a week after
Defining optimal methadone dosing in pregnancy has delivery. Patients who manifested recurrent withdrawal symp-
been difficult due to a lack of pharmacokinetic information to toms requiring repeated dose increases had levels drawn more
guide dosing (Jeong, 2010). In a study of the newly appre- often as part of individualized patient care. The data were
ciated primary role of CYP2B6 in methadone metabolism, further collectively monitored as part of internal program
Amunugama et al. (2012) conclude that: ‘‘Our understanding quality assurance. Serum level and ratio data were discussed
of the metabolism of methadone is incomplete; our ability to in both group and individual settings to assist mothers in
accurately predict pharmacokinetics in individuals leads to understanding their unique metabolism and the implications
adverse effects, to significant drug–drug interactions, and to a for fetal exposure and individualized dosing.
number of fatalities.’’ Further, the unpredictability of meth- Dose regimens were adjusted clinically based on patient
adone’s effects at a specific dose results in a high incidence of assessment and on symptoms reported on the Subjective Opiate
over and underdosing which can cause severe adverse events Withdrawal Scale (SOWS) augmented for pregnancy (McCar-
such as withdrawal symptoms, respiratory depression, and thy et al., 2015). Using 600 ng/mL as the upper limit for
potential QTc prolongation that can result in sudden death (Lu methadone trough serum concentration, dose and dose fre-
et al., 2010). quency were increased to minimize emergent maternal with-
Serum methadone levels provide an objective measure drawal symptoms. Because single doses of methadone are
of metabolism, and, indirectly, of fetal exposure. Although a associated with physiologic abnormalities in the fetus (Whitt-
relationship between methadone serum concentration and mann and Segal, 1991; Jansson et al., 2009), all pregnant
therapeutic effect is not precisely defined (Shiu and Ensom, patients were transitioned to a twice daily regimen (2 equally
2012), a concentration range of 200 to 600 ng/mL has long divided doses) within a few days of admission, pending regu-
served as a clinical guide to effective treatment (Dole, 1988), latory approval for a daily take home dose. Our divided dosing
and this range has been found to be a clinically safe and protocol is described elsewhere (McCarthy et al., 2015). Most
effective range for pregnant patients (McCarthy et al., 2015). patients were on 3 to 4 times a day dosing at the time of delivery,
Peak and trough serum concentration ratios (PTRs) (the although the regimen varied between 2 and 6 times daily.
trough before the morning dose and the peak 4 hours later) Postpartum serum levels were drawn upon return to the clinic
have traditionally been used to document hypermetabolism, after delivery (usually 3–7 days postpartum), and repeated in 2
with a ratio greater than 2 indicating more rapid clearance to 3 weeks. Most patients had dose reductions in the early
than the usual 24-hour norm (Westermeyer et al., 2016). postpartum period, with patients delivering on higher doses
However, requiring 2 blood draws 4 hours apart on the same undergoing more rapid dose reductions.
day poses a prohibitive logistical barrier for many patients. A All serum levels were drawn at the clinic just before the
single blood test measuring methadone, the EDDP metabolite, morning dose and sent to San Diego Reference Laboratory.
and their calculated ratio (methadone/metabolite ratio Measurements for methadone and metabolite were done using
[MMR]) is more practical and provides a more dynamic high pressure liquid chromatograph/triple quadrupole mass
picture of metabolism than simple serum levels, with the spectrometer (HPLC/MSS/MS). The data for comparing dif-
additional ability to categorize individuals based on their ferent trimesters contain a mix of paired and unpaired data.
phenotypic ratios stratified into poor metabolizers (PMs, ratio Our analysis used the permutation test of Einsporn and
greater than 16), intermediate metabolizers (IMs, ratio 12– Habtzghi (2013) with 5000 replications. When a patient
15), extensive metabolizers (EMs, ratio 5–11), and ultrarapid had several measurements in a period, this analysis used
metabolizers (URMs, ratio 4 or less) (Zanger and Schwab, the mean for that period. All significant results were con-
2013). The MMR gives information on the net effect of the firmed with t tests using only paired or independent parts of
multiple enzymes involved in methadone metabolism. A the data depending on the greatest degrees of freedom.
lower ratio indicates more rapid metabolism, and accordingly Calculations were made using R version 3.4.1. Reported
less opioid activity at a given dose. P values are 2-tailed.

2 ß 2018 American Society of Addiction Medicine

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.
CE: M.S.; JAM-D-17-00166; Total nos of Pages: 6;
JAM-D-17-00166

J Addict Med  Volume 00, Number 00, Month/Month 2018 Monitoring Changing Perinatal Pharmacokinetics

TABLE 1. Dose, Serum, Metabolite, and Ratio Means and Ranges


Dose (Range), mg/d Serum (Range), ng/mL Metabolite (Range), ng/mL M/M Ratio (Range)
Prepartum 154 (35–355) 288 (78–854) 50 (16.9–142.3) 5.9 (2.6–11.6)
Postpartum 205 (95–340) 434 (169–1020) 66 (21–231.8) 7.2 (3.7–19.8)

Means use within period patient averages.

The UC Davis IRB Administration reviewed this study individual trends over time, data from 4 patients with the most
and determined it was not research involving human subjects pre/postpartum tests results are shown in Table 2.
(FWA No: 00004557). Prepartum, methadone serum level and dose were posi-
tively correlated (correlation ¼ 0.53, P < 0.0001), as were
RESULTS serum level and metabolic ratio (correlation ¼ 0.41,
Between January, 2015 and January, 2016, 23 patients P ¼ 0.002). Dose and ratio were negatively correlated, but
provided 67 levels during pregnancy (13 from first trimester, not significantly (correlation ¼ 0.15, P ¼ 0.29). Ratios
30 from second, 24 from third), and 11 of these patients changed significantly over time. Table 3 shows average
provided 22 postpartum levels. Sixteen patients (70%) con- within-patient ratios decreasing from 7.2 in the first trimester,
ceived on methadone and 7 (30%) started treatment who were to 5.9 in the second trimester, to 5.1 in the third trimester, and
already pregnant. then increasing to 7.2 postpartum. The ratios declined signifi-
Prepartum, the average methadone dose for the cohort cantly from trimester 1 to trimester 3 (P ¼ 0.007). Ratios then
was 154 mg/d; the average serum level was 288 ng/mL; the rose rapidly and significantly from trimester 3 to the postpartum
average metabolite level was 50 ng/mL; and the average ratio period (P ¼ 0.001), reaching levels similar to those of trimester
was 5.9. Postpartum, the average dose was 205 mg/d; the 1. There was a highly significant increase in average serum
average serum level was 434 ng/mL; the average EDDP levels from third trimester to postpartum (P ¼ 0.001). Four of
metabolite level was 66 ng/mL; and the average ratio was 13 patients (31%) had serum levels that exceeded the therapeu-
7.2 (see Table 1 for additional summary data). To illustrate tic range of 600 ng/mL, 1 going as high as 1020 ng/mL.

TABLE 2. Individual Trends in Methadone Dose, Metabolite, and Ratio (Example Data)
Trimester/ Methadone Methadone Serum EDDP Serum Methadone/
Patient ID Postpartum Daily Dose, mgy Level, ng/mL Level, ng/mL EDDP Ratio
15 T2 220 251 48 5.2
T3 280 365 88 4
T3 325 370 94 3.9
T3 355 361 84 3.8
PP1 340 672 89 7.6
PP2 300 591 61 9.7
PP3 300 756 56 13.5
PP4 140 494 25 19.8
16 T1 200 435 82 5.3
T2 260 340 89 3.8
T2 270 447 118 3.8
T2 280 469 72 6.5
T3 280 516 91 5.7
T3 310 854 143 6
PP1 260 1020 230 4.4
PP2 240 987 156 6.3
PP3 200 909 126 7.2
17 T2 125 195 44 3.4
T2 135 125 37 3.4
T3 190 129 33 3.9
PP1 200 206 35 5.9
PP2 190 192 27 7.1
23 T2 160 213 53 4
T3 210 294 30 8.6
T3 260 315 96 3.3
T3 280 321 80 4
T3 310 310 118 2.6
PP1 280 727 131 5.5
PP2 240 467 55 8.5
PP, postpartum; T, trimester.

Listed chronologically, time intervals between samples varied based on clinical need. All postpartum levels were within the first month postpartum.
yTotal daily dose, divided into 2 or more separate doses across the day.

ß 2018 American Society of Addiction Medicine 3

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.
CE: M.S.; JAM-D-17-00166; Total nos of Pages: 6;
JAM-D-17-00166

McCarthy et al. J Addict Med  Volume 00, Number 00, Month/Month 2018

TABLE 3. Methadone/Metabolite (EDDP) Ratio by as failure to identify such patients and increase doses and
Trimester dosing frequencies will likely result in significant
maternal withdrawal.
Mean Median N (Individual
Trimester Ratio Ratio SD Patients)
Methadone/metabolite ratios have been the subject of
toxicological studies in patient populations. A study of 32
1 7.2 7.2 1.9 11 nonpregnant methadone maintenance patients measured
2 5.9 5.2 2.7 16
3 5.1 4.9 1.3 16 ratios at both peak and trough, and found mean ratios virtually
PP 7.2 6.5 3.3 11 identical: peak mean 11.3 and trough mean 12.7 (Diong et al.,
PP, postpartum.
2014). Metabolic ratios, unlike serum levels, are constant

Data using each patient’s average ratio per trimester. through a 24-hour period. Another toxicological study of
MMRs in 23 nonpregnant patients found a mean ratio of
11.9 (Alburges et al., 1996). Our ratio data found a mean
The percent of ratios that were 4 or less, indicating of 6.3 for all patients (pregnant and postpartum), and a mean
ultrarapid metabolism, increased from 8% (n ¼ 1/13) in the of 6.1 for all actively pregnant patients. Furthermore, in our
first trimester, to 30% (n ¼ 9/30) in the second, to 38% (n ¼ 9/ population, the average first trimester ratio was 7.2, indicating
24) in the third, and decreased to 5% (n ¼ 1/22) postpartum. that even in the first trimester, pregnant patients had ratios
Forty-four per cent (n ¼ 10/23) of individual patients had at well below the ratios found in nonpregnant samples.
least 1 prepartum ratio of 4 or less. The number of ultrarapid A study of peak/trough methadone serum ratios (PTRs)
metabolizers by trimester was n ¼ 1 (9%) in trimester 1, n ¼ 7 in an all-male population found a strong effect of dose on peak
(44%) in trimester 2, n ¼ 5 (31%) in trimester 3, and n ¼ 1 serum level, but a weak effect on trough level (Westermeyer
(9%) postpartum. et al., 2016). This illustrates the important point that, once
adequate peak level has been achieved, merely increasing the
DISCUSSION dose in rapid metabolizing patients (ie, with a PTR of greater
This is the first report, to our knowledge, of the use of than 2) would unnecessarily increase the peak, while having
methadone/metabolite serum ratios to monitor metabolic minimal effect on the trough. This is especially important in
changes in the perinatal period, providing information on pregnancy. Merely increasing doses to treat emergent with-
the net effect that the various P450 enzyme changes induced drawal in pregnant patients exposes the fetus to higher peak
by pregnancy have on methadone metabolism. Our use of levels without effecting the trough, resulting in potential
sequential ratios allowed measurement of a significant adverse effects on fetal physiology which is sensitive to
decrease in ratios from first to third trimesters, and signifi- peak/trough extremes. Achieving pharmacodynamic stability
cantly increased ratio from third trimester to postpartum. This at the fetal mu receptor in the context of dramatically altered
clinical data support the CYP enzyme induction study of maternal methadone pharmacokinetics requires altering both
dextromethorphan metabolism by Tracy et al. (2005), who the dose and the dose regimen to compensate for the reduced
found that CYP2D6 activity was increased throughout the half-life of the medication. Westermeyer et al. (2016) con-
pregnancy (25% at 14–18 weeks, 35% at 24–28 weeks, and clude that greater use of serum levels can ‘‘help clinician/
48% at 36–40 weeks), and CYP3A was consistently and patient dyads collaborate in detecting inadequate doses and
significantly increased (35–38%) during all stages of preg- flawed dosing strategies, thereby preventing needless with-
nancy. Our data document accelerated metabolism through all drawal symptoms, craving, and recurrent addiction.’’
phases of pregnancy and do not support the concept that The use of PTRs and our use of MMRs are different
accelerated metabolism and need for dose changes are only a ways of monitoring the same metabolic process, but use of
third trimester event. In some patients, the lowest ratios were MMRs has the significant advantage of requiring only a single
in the second trimester. In 1 patient, serum level and ratio blood draw, which can even be done using a finger stick
unexpectedly increased before delivery, indicating that the without the need for sometimes difficult phlebotomies. PTRs
accelerated metabolism of pregnancy, although generally also lack the element of standardization achieved through the
increasing during pregnancy, can fluctuate, illustrating the 4 established categories of metabolism (slow, intermediate,
importance of close serum level/ratio monitoring. Different extensive, and ultrarapid). Furthermore, there is the potential
CYP450 enzymes appear to be induced at different points in to be explored of extrapolating from MMRs to an estimation
time and likely at different levels of expression all through of half-life of methadone, a concept more familiar to physi-
the pregnancy. cians, since ratios are a surrogate for half-life. MMRs, then,
Our data indicate that 44% of the patients, at some point are an advance over PTRs in terms of ease of use and
in the pregnancy, show ratio evidence of ultrarapid metabo- metabolic information, and their use should promote better
lism (URM), and that 44% and 31% of patients in second and physician understanding and more effective of dosing of
third trimesters, respectively, have evidence of URM. This is mother and fetus.
much higher than known baseline rates of URM in nonpreg- Postpartum ratios increased rapidly by 41% over third
nant populations. Diong et al. (2014) found that 15% of a trimester values, making the postpartum period, arguably, the
nonpregnant population had a peak/trough ratio greater than 2, most dramatic period of pharmacokinetic change in adult
indicating URM by that measure. URM has been associated human physiology. Monitoring postpartum serum levels and
with poor response to treatment (Eap et al., 2001). The very ratios are important for patient safety, as escalating serum
high rate of URM in pregnant patients raises special concerns, levels can cause oversedation and impair mothering. Dose

4 ß 2018 American Society of Addiction Medicine

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.
CE: M.S.; JAM-D-17-00166; Total nos of Pages: 6;
JAM-D-17-00166

J Addict Med  Volume 00, Number 00, Month/Month 2018 Monitoring Changing Perinatal Pharmacokinetics

reductions should occur in most patients in the immediate While this pilot study focused on pregnancy as an
postpartum period, especially those requiring higher doses. inducer of methadone metabolism, the MMRs may have
Further, it is important that the mothers not become dependent important use in other clinical situations. Foremost would
on high, clinically unnecessary, serum levels. There is evi- be the potential use of MMRs to monitor drug–drug inter-
dence from a study of midazolam (a 3A4 substrate) metabo- actions, such as occurs with co-administration of some anti-
lism in pregnancy that the return to a prepartum metabolic depressants, anticonvulsants, and antifungals. The effect of
state may take longer than 10 weeks, making serial serum these interactions on methadone concentration is poorly
level monitoring advisable (Hebert et al., 2008). Serum levels predicted by current studies. Further, there is not yet full
and ratios after delivery provide a clear measure of individual consensus on the involvement of the various CYP450
changes to guide safe dosing, especially when late third enzymes known to metabolize methadone (Levran et al.,
trimester levels are used for comparison. 2013), and it has been suggested that guidelines warning of
Dose increases in our study were significantly corre- CYP3A4-mediated drug interactions may be incorrect (Khar-
lated with serum level increases, indicating that increases asch et al., 2009). The metabolic ratio, representing the net
required to eliminate withdrawal symptoms are not just effect of whatever CYP enzymes are involved in a particular
returning the patient serum level to the previous baseline. patient, could provide a quantitative alert to the clinician of
It suggests that there are other factors that affect drug dispo- the effect of such medications on methadone metabolism, if a
sition and efficacy in pregnancy. Permeability glycoprotein baseline ratio is obtained before starting a new medication.
(P-gp) is a cell membrane protein that actively transports Further, metabolic changes can occur as a result of illness (eg,
drugs out of the cell, reducing drug levels at the target site (Lin renal or hepatic disease), and the MMR could be used to
and Yamazaki, 2003). Methadone is a P-gp substrate and its document such changes. One could even make a case for
activity is increased during pregnancy (Rollason et al., 2008). establishing a baseline ratio on all patients treated with
P-gp would be expected to decrease the efficacy of methadone methadone. The MMR is a unique identifier for an individual
serum concentrations by actively transporting methadone patient, based on his/her genetic polymorphisms for the
away from the mu receptor. Further, CYP450 enzymes have enzymes that are involved with the kinetic conversion of
been found to have brain-specific expression that may differ methadone. Ratios not only identify URM who are at risk
from hepatic forms, such that patient response may not be for poor treatment response, but also poor metabolizers who
predicted by serum levels (Levran et al., 2013). These factors are at risk for unusually high serum levels at routine doses
are consistent with the range of serum levels in our study with potential for sedation, overdose (Richards-Waugh et al.,
(range 58–854 ng/mL) that were associated with, at least 2014), and arrhythmias (Wadelius et al., 1997).
temporary, clinical stability. The goal of individualized medicine is for physicians to
Three studies have found evidence of increased metha- have the information necessary to prescribe an appropriate
done clearance during pregnancy: studies by Pond et al. (1985), medicine to the right target of the disease at the right dose and
Jarvis et al. (1999), and Wolff et al. (2005). However, metha- dose frequency for individual patients to achieve maximal
done concentration is influenced by pharmacokinetic factors benefit with minimal side effects (Ma and Lu, 2011). Serum
beyond CYP450 metabolism. Jarvis et al. (1999) found evi- levels and metabolic ratios improve the quality of information
dence of decreased absorption of methadone in pregnancy, physicians need to achieve these goals in the case of pregnant
which would decrease concentration but would not alter the patients on methadone. In the future, patients could be
MMR. Pregnancy can increase renal clearance of drugs (Jeong, genotyped for their response to methadone. At this time,
2010). However, although clearance of both total and unbound however, a simple, inexpensive blood test can give important
methadone is greater during pregnancy, renal clearance was clinical information on the net effect of both genetics and
found to contribute only minimally to methadone clearance pregnancy on stability of maternal, and also fetal, methadone
(Pond et al., 1985), at least at normal urinary pH (Diong et al., exposure. The pregnant patient and her fetus deserve scien-
2014). Furthermore, the percentage of methadone dose tifically based dosing and close physician monitoring as a
excreted as methadone and its metabolites did not change standard of care if we are to optimize outcomes of
during pregnancy, that is, there was no change that would alter these pregnancies.
their metabolic ratio (Pond et al., 1985). Blood volume expan-
sion in pregnancy reduces concentration of albumen and alpha REFERENCES
1 glycoprotein, leading to decreased binding and a potential Alburges ME, Huang W, Foltz RL, et al. Determination of methadone and its
increase in unbound drug (Jeong, 2010). While protein binding N-demethylation metabolites in biological specimens by GC-PICI-MS.
was found to be less early in pregnancy than postpartum (Pond J Anal Toxicol 1996;20:362–368.
et al., 1985), any increase in unbound methadone would be Amunugama HT, Zhang H, Hollenberg PF. Methadone-based inactivation of
cytochrome P450 2B6 by methadone through destruction of prosthetic
offset by the greater clearance of both bound and unbound drug. Heme. Drug Metabol Dispos 2012;40:1765–1770.
Lehotay et al. (2005) found that the R-form (the active enan- Bogen DL, Perel JM, Helsel JC, et al. Pharmacologic evidence to support
tiomer) MMR was the same regardless of dose, so different clinical decision making for peripartum methadone treatment. Psycho-
doses would not affect the MMR. In summary, while serum pharmacology 2013;225:441–451. doi:10.1007/s00213-012-2833-7.
concentration could be affected by various physiologic Corr TE, Hollenbeak CS. The economic burden of neonatal abstinence
syndrome in the United States. Addiction 2017;112:1590–1599.
changes, the MMR is independent of these factors and is Dickmann LJ, Isoherranen N. Quantitative prediction of CYP2B6 induction
therefore an adequate unitary measure of methadone metabo- by estradiol during pregnancy: potential explanation for increased metha-
lism during pregnancy and post-partum. done clearance during pregnancy. Drug Metab Dispos 2013;41:270–274.

ß 2018 American Society of Addiction Medicine 5

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.
CE: M.S.; JAM-D-17-00166; Total nos of Pages: 6;
JAM-D-17-00166

McCarthy et al. J Addict Med  Volume 00, Number 00, Month/Month 2018

Diong SH, Yusoff NS, Sim MS, et al. Quantitation of methadone and Ma Q, Lu AYH. Pharmacogenetics, pharmacogenomics, and individualized
metabolite in patients under maintenance treatment. J Anal Toxicol medicine. Pharmacol Rev 2011;63:437–459.
2014;38:660–666. Mattick RP, Breen C, Kimber J, et al. Buprenorphine maintenance versus
Dole V. Implications of methadone maintenance for theories of narcotic placebo or methadone maintenance for opioid dependence. Cochrane
addiction. JAMA 1988;260:3025–3029. Database Syst Rev 2014;CD002207.
Eap CB, Broly F, Mino A, et al. Cytochrome P450 2D6 genotype and McCarthy JJ, Leamon MH, Willits NH, et al. The effects of methadone dose
methadone steady-state concentrations. J Clin Psychopharmacol 2001; regimen on neonatal abstinence syndrome. J Addict Med 2015;9:
21:229–234. 105–110.
Eap CB, Boclin T, Baumann P. Interindividual variability of the clinical McCarthy JJ, Leamon MH, Finnegan LP, et al. Opioid dependence and
pharmacokinetics of methadone: implications for the treatment of opioid pregnancy: minimizing stress on the fetal brain. Am J Obstet Gynecol
dependence. Clin Pharmacokinet 2002;41:1153–1193. 2017;216:226–231.
Einsporn RL, Habtzghi D. Combining paired and two-sample data using a Nebert DW, Nelson DR, Feyereisen R. Evolution of the cytochrome P450
permutation test. J Data Science 2013;11:767–779. genes. Xenobiotica 1989;19:1149–1160.
Hebert MF, Easterling TR, Kirby B, et al. Effects of pregnancy on 3A4 and P- Pond SM, Kreek MJ, Tong TG, et al. Altered methadone pharmacokinetics in
glycoprotein activities as measured by disposition of midazolam and methadone-maintained pregnant women. J Pharmacol Exp Ther 1985;
dogoxin. Clin Pharmacol Ther 2008;84:248–253. 233:1–6.
Isoherranen N, Thummel KE. Drug metabolism and transport during pregnancy: Richards-Waugh LL, Primerano DA, Dementieva Y, et al. Fatal methadone
how does drug disposition change during pregnancy and what are the toxicity: potential role of CYP3A4 genetic polymorphism. J Analyt Toxicol
mechanisms that cause such changes. Drug Metab Dispos 2013;41:256–262. 2014;38:541–547.
Jansson LM, DiPietro JA, Velez M, et al. Maternal methadone dosing Rollason V, Samer C, Piguet V, et al. Pharmacogenetics of analgesics: toward
schedule and fetal neurobehavior. J Matern-Fetal Neonatal Med 2009; the individualization of prescription. Pharmacogenomics 2008;9:
22:29–35. 905–933.
Jarvis MAE, Wu-Pong S, Kniseley JS, et al. Alterations in methadone Shiu JR, Ensom MHH. Dosing and monitoring of methadone in pregnancy:
metabolism during late pregnancy. J Addict Dis 1999;18:51–61. literature review. Can J Hosp Pharm 2012;65:380–386.
Jeong H. Altered drug metabolism during pregnancy: hormonal regulation of Tracy TS, Venkataramanan R, Glover DD, et al. Temporal changes in drug
drug-metabolizing enzymes. Expert Opin Drug Metab Toxicol 2010; metabolism (CYP1A2, CYP2D6 and CYP3A activity) during pregnancy.
6:689–699. Am J Obstet Gynecol 2005;192:633–639.
Kharasch ED, Hoffer C, Whittington D, et al. Methadone pharmacokinetics Wadelius M, Darj E, Frenne G, et al. Induction of CYP2D6 in pregnancy. Clin
are independent of cytochrome P4503A (CYP3A) activity and gastroin- Pharmacol Ther 1997;62:400–407.
testinal drug transport: insights from methadone interactions with ritona- Weinshilboum R. Inheritance and drug response. N Engl J Med 2003;
vir/indinavir. Anesthesiology 2009;3:660–672. 348:529–537.
Lehotay D, George S, Etter M, et al. Free and bound enantiomers of Westermeyer J, Yoon G, Thuras P, et al. Pharmacotherapy in methadone
methadone and its metabolite, EDDP in methadone maintenance treat- maintenance: clinical utility of peak-trough blood levels. Addict Disord
ment: relationship to dosage? Clin Biochem 2005;38:1088–1094. Their Treatment 2016;15:157–164.
Levran O, Peles E, Harmon S, et al. CYP2B6 SNPs are associated with Whittmann BK, Segal S. A comparison of the effects of single- and split dose
methadone dose required for effective treatment of opioid addiction. methadone administration on the fetus: an ultrasound evaluation. Int J
Addict Biol 2013;18:709–716. Addict 1991;26:213–218.
Lin JH, Yamazaki M. Role of P-glycoprotein in pharmacokinetics: clinical Wolff K, Boys A, Rostami-Hodjegan. et al. Changes to methadone clearance
implications. Clin Pharmacokinet 2003;42:59–98. during pregnancy. Eur J Clin Pharmacol 2005;61:763–768.
Lu WJ, Bies R, Kamden LK, et al. Methadone: a substrate and mechanism- Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism:
based inhibitor of CYP19 (aromatase). Drug Metab Dispos 2010;38: regulation of gene expression, enzyme activities, and impact of genetic
1308–1313. variation. Pharmacol Ther 2013;138:103–141.

6 ß 2018 American Society of Addiction Medicine

Copyright © 2018 American Society of Addiction Medicine. Unauthorized reproduction of this article is prohibited.

You might also like