Dose-adjusted plasma concentrations of sublingual buprenorphine are lower during than after pregnancy




Background


Buprenorphine is a Food and Drug Administration–approved maintenance therapy for opioid use disorders and is increasingly being used in pregnant women with opioid use disorders as an alternative to methadone. Dosing of buprenorphine in pregnant women is based on the regimen recommended for nonpregnant females and males. Limited data are available defining the pharmacokinetic properties of sublingual buprenorphine administered during pregnancy.


Objective


This study evaluated the impact of physiological changes associated with pregnancy on the pharmacokinetics of sublingual buprenorphine during and after pregnancy.


Study Design


Pregnant women (n = 13), between 18 0/7 and 37 6/7 weeks’ singleton gestation, receiving sublingual buprenorphine twice daily for opioid use disorders were studied. Pharmacokinetic-2 studies were performed between 18 and 25 weeks (n = 7), pharmacokinetic-3 studies were performed between 31 and 37 weeks (n = 11), and pharmacokinetic-P was performed 4-18 weeks postpartum (n = 10). On the day of the study, blood was withdrawn prior to the daily morning dose of buprenorphine and at 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 8, and 12 hours after the dose. Buprenorphine plasma concentrations were analyzed by liquid chromatography tandem mass spectrometric detection. All pharmacokinetic parameters were observed or estimated using Microsoft Excel. Statistical analyses were performed to identify significant changes in study participants’ buprenorphine pharmacokinetic parameter estimates over the duration of the study. Univariate linear and generalized linear mixed models were used to investigate changes in these measures over time, some of which were log transformed for normality.


Results


Dose-normalized (plasma concentration per dose) buprenorphine plasma concentrations were significantly lower during pregnancy (pharmacokinetic-2 plus pharmacokinetic-3) than during the postpartum period (pharmacokinetic-P). Specific pharmacokinetic parameters (and level of significance) were as follows: the area under the buprenorphine plasma concentration-time curves ( P < .003), maximum buprenorphine concentrations ( P < .018), buprenorphine concentrations at 0 hour ( P < .002), and buprenorphine concentrations at 12 hours ( P < .001). None of these parameters differed significantly during pregnancy (ie, pharmacokinetic-2 vs pharmacokinetic-3). The time to maximum buprenorphine concentrations did not differ significantly between groups.


Conclusion


The dose-normalized plasma concentrations during a dosing interval and the overall exposure of buprenorphine (area under the buprenorphine plasma concentration-time curves) are lower throughout pregnancy compared with the postpartum period. This indicates an increase in apparent clearance of buprenorphine during pregnancy. These data suggest that pregnant women may need a higher dose of sublingual buprenorphine compared with postpartum individuals. The dose of buprenorphine should be assessed after delivery to maintain similar buprenorphine exposure during the postpartum period.


Opioid use disorders in America have increased at an alarming rate during the past decade. Among pregnant women aged 15–44 years, 5.4% admitted to currently using illicit drugs, with the highest rates during the first (9%) and second (4.8%) trimester compared with the third (2.4%) trimester.


Untreated substance use during pregnancy not only increases maternal pregnancy complications but also increases fetal risk. Specifically, untreated chronic heroin use is associated with an increased risk of pregnancy complications, such as fetal growth restriction, placental abruption, fetal death, preterm labor, third-trimester bleeding, fetal distress, meconium aspiration, and puerperal morbidity.


Currently methadone is most often prescribed as first-line pharmacotherapy for a pregnant woman with opioid use disorders, but buprenorphine use has increased, as recent evidence suggests comparable efficacy and less severe neonatal complications with buprenorphine compared with methadone. Dosing of buprenorphine is based mostly on expert panel recommendations and subjective data collected from the patient and is dose adjusted using patient symptoms of withdrawal.


Despite such recommendations, there is a lack of consensus concerning appropriate induction and maintenance dosing, monitoring parameters, and duration of therapy because of a wide range of clinical and contextual factors and concerns about diversion. Buprenorphine is extremely lipophilic, highly bound to plasma proteins, and mainly metabolized by cytochrome P450 enzyme superfamily (CYP3A4) and glucuronosyltransferase enzyme superfamily (UGT1A/2B). Pregnancy can substantially alter drug absorption, distribution, metabolism, and/or elimination, possibly leading to changes in the most effective dose or dosing regimen that should be used in this specific patient population.


The current investigation examined the pharmacokinetics (PK) of buprenorphine administered sublingually during and after pregnancy to determine whether there are differences in the PK estimates during pregnancy as well as between pregnancy and the postpartum period.


Materials and Methods


Participants


Women (n = 17) were recruited from Magee-Womens Hospital (Pittsburgh, PA) and an outlying clinic. Participants were recruited and enrolled from June 1, 2014, through Nov. 30, 2014. All participants were receiving twice-daily buprenorphine maintenance therapy, as prescribed for clinical purposes by their respective caregivers, and were expected to be at steady state on their current dose prior to each study visit. The protocol was approved by the University of Pittsburgh’s Institutional Review Board, and all participants underwent the informed consent process using institutional review board-approved consent documents.


Procedures


Demographic details, baseline laboratory parameters, medication, and/or substance use history and obstetric history were collected for all participants. Eligibility criteria for these women included the following: (1) pregnant and on a stable, twice-daily dose of buprenorphine for at least 7 days, (2) ≥18 years of age, (3) able to willingly consent, and (4) willing to have urine samples screened for the presence of alcohol, barbiturates, opiates, cocaine (or metabolites), benzodiazepines, synthetic opioids, PCP, and concurrent medications or substances.


Exclusion criteria included the following: (1) hepatic or renal dysfunction, (2) sickle-cell disease and on active treatment, (3) HIV and on active treatment (potential drug interactions), (4) hypersensitivity to opioids, (5) comorbid dependence on benzodiazepines, (6) concurrently taking monoamine oxidase inhibitors, neuroleptics, or disulfiram, and (7) concurrently taking medications known to be CYP3A inducers (such as rifampin, phenobarbital, phenytoin, or carbamazepine) or CYP3A inhibitors (such as azole antifungals, macrolide antibiotics, or HIV protease inhibitors).


Up to 3 studies were performed in each participant. PK-2 studies were performed in the second trimester between 18 and 25 weeks (n = 7), PK-3 studies were performed in the third trimester between 31 and 37 weeks (n = 11), and PK-P was performed 4-18 weeks postpartum (n = 10). All PK study visits were identical and conducted in the Clinical and Translational Research Center of the University of Pittsburgh Medical Center’s Montefiore Hospital.


Participants arrived early in the morning after a ≥8 hour fast with their medication log, underwent a predose blood draw (BD Vacutainer glass blood collection tubes with sodium heparin; Becton Dickson Inc, Franklin Lakes, NJ) and oral fluid collection (Sarstedt salivette cotton swab for saliva collection; Sarstedt Inc, Newton, NC) for trough buprenorphine concentrations and clinical laboratory parameters; provided a urine sample for laboratory tests and toxicology; had oral pH recorded (Hydrion urine and saliva pH paper, range 5.5–8.0; Micro Essential Lab, Brooklyn, NY); and then took their prescribed sublingual buprenorphine dose under direct supervision by study staff.


Participants were instructed to allow the tablets to dissolve under their tongue, with minimal swallowing, until there was no visible residue remaining. Dissolution time was recorded after visual inspection of the sublingual area by study staff. Serial blood and oral fluid samples were subsequently collected over 1 dosing interval at 0 (before the dose), 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 8, 10, and 12 hours after the dose. All blood samples and oral fluid salivettes were centrifuged for 15 minutes at 15,000 rpm to obtain plasma and oral fluid, respectively. Samples were immediately frozen at –80°C until analysis.


All spontaneously voided urine was collected throughout the entire study period of 12 hours. Participants were not allowed to eat until 2 hours after the dose to allow for adequate drug absorption, and all food consumed during the study day was reported on a dietary log.


Assay methodology


Buprenorphine (BUP) and its 3 active metabolites (norbuprenorphine, buprenorphine glucuronide, and norbuprenorphine glucuronide), along with the deuterated internal standards for BUP, norbuprenorphine, and norbuprenorphine glucuronide (buprenorphine glucuronide was not available), were extracted from plasma samples by solid-phase extraction methods.


Plasma concentrations were determined using high-performance liquid chromatography with tandem mass spectrometric detection. The peaks of interest were well separated and the overall run time for each sample was 7 minutes. Detection was accomplished utilizing ion spray tandem mass spectrometry in positive ion multiple reaction monitoring mode. The lower limit of quantification was 0.05 ng/mL for BUP, and calibration curves were linear, ranging from 0.05 to 50 ng/mL for BUP with coefficients of determination (r2) greater than 0.99.


Both the intraday and interday precisions were evaluated and the coefficient of variation values were less than 15% for low, medium, and high controls. The accuracy as measured by bias was less than 5%. Cumulative urine and serial oral fluid samples were also collected and concentrations measured (data not shown).


Statistical analysis


Noncompartmental analysis was performed and various PK parameters were calculated using Microsoft Excel (Richmond, WA). Maximum BUP plasma concentrations (Cmax), trough BUP concentrations at time zero (C0), BUP concentrations at 12 hours (C12), and time to maximum BUP concentrations (Tmax) were observed values using each participant’s plasma concentration-time profile.


In each of the participants, the area under the BUP plasma concentration-time curve for the 12 hour dosing interval (AUC 0→12 ) was calculated from time 0 to 12 hours using the trapezoidal rule. The mean values were then used to compare cohorts. Given the reportedly long half-life of buprenorphine (approximately 37 hours) and the shorter dosing interval (12 hours) used in the study participants, it was not possible to calculate the terminal half-life.


Univariate descriptive statistics were used to summarize sample demographic and clinical characteristics at each study visit. Means and standard deviations were reported for continuous variables, and frequencies and percentages were reported for categorical variables. BUP pharmacokinetic data were similarly summarized descriptively at each study visit using means and standard deviations. Statistical analyses were then performed to identify significant differences in BUP PK parameters between (1) PK-2 and PK-3 and (2) pregnancy and postpartum, without focus on a specific trimester of pregnancy.


The distributions of sample PK data were investigated, and a natural logarithmic transformation was applied to each of the PK variables to produce approximately normally distributed data for statistical analysis. Univariate linear mixed models were then fit to the log-transformed PK data to make the 2 comparisons of interest because such modeling techniques utilize data from every participant and do not require that participants have complete data at all 3 study visits.


Time (PK-2, PK-3, and PK-P) was treated as a categorical fixed effect in all models, and a random effect was included in the models to account for repeated measures over time from the same participants. For the random effect of time, a variance component covariance structure was assumed. Models were fit through maximum likelihood estimation in SAS version 9.4 (SAS Institute, Cary, NC), and convergence criteria were met without problems.


The fitted models were then used to compare PK parameters between the second and third trimesters as well as between pregnancy and postpartum, using Student t tests. Specifically, the first comparison tested whether the log-transformed PK parameter differed significantly between PK-2 and PK-3. For the second comparison, a formal hypothesis test was performed to determine whether the log-transformed mean at PK-P differed significantly from the average of the log-transformed means at PK-2 and PK-3. The 2 P values were adjusted for multiple comparisons using the Sidak adjustment method. A value of P < .05 indicated a statistically significant difference.




Results


Seventeen participants provided informed consent for the study. Two participants were excluded after baseline assessment: one for a failed urine comprehensive drug screen and one for an ectopic pregnancy. One participant was lost to follow-up after consent and was never screened. Thus, data from 14 pregnant women were available for pharmacokinetic analysis, and 6 of these 14 participants were studied at all 3 time points.


Among these 14 participants, a total of 35 pharmacokinetic studies were completed: 9 in PK-2, 13 in PK-3, and 13 in PK-P. Of these 35 studies, 7 were deemed invalid: 2 participants’ data were excluded from the PK-2 analysis, and 2 participants’ data were excluded from PK-3 analysis because the buprenorphine concentration at time 0 was significantly higher than the buprenorphine concentration at 12 hours, indicating participants had an atypical concentration-time profile and were likely not adherent to a 12 hour dosing interval.


Data from 2 participants were excluded from the PK-P analysis because they were switched by their providers to Suboxone film (buprenorphine/naloxone) at the time of the study, which may have different pharmacokinetic characteristics than sublingual buprenorphine tablets. Data from 1 participant was also excluded from the PK-P analysis after her medication log showed once-daily rather than twice-daily buprenorphine dosing.


This resulted in 28 studies with pharmacokinetic data being available for further analysis (18 from the 6 participants who were studied at all 3 time points): 7 for PK-2, 11 for PK-3, and 10 for PK-P.


The characteristics of the study participants are listed in Table 1 according to the time of study. The mean gestational ages were 22.0 weeks for PK-2 and 33.9 weeks for PK-3. The PK-P study was performed at a mean of 7.4 weeks after delivery. All but 1 participant studied postpartum (PK-P) was also studied at least once in pregnancy (PK-2 or PK-3 or both).



Table 1

Characteristics of study participants
































































Characteristic (mean ± SD) PK-2 (second trimester) (n = 7) PK-3 (third trimester) (n = 11) PK-P (postpartum) (n = 10)
Age, y 27.3 ± 5.0 27.9 ± 5.2 28.6 ± 5.6
Smoker, current 5 (71.4%) 8 (72.7%) 7 (70.0%)
Parity, nulliparous 2 (28.6%) 3 (27.3%) 2 (20.0%)
Twice-daily dose, mg 8.0 ± 3.1; range, 2–12 10.0 ± 3.7; range, 2–16 8.0 ± 0.0
Gestational age, wks 22.0 ± 2.5 34.0 ± 2.2
Postpartum, wks 7.1 ± 4.2
Body weight, kg 73.7 ± 7.6 74.1 ± 8.3 66.5 ± 9.4
BMI, kg/m 2 28.5 ± 3.8 28.6 ± 3.9 25.3 ± 4.6
Waist circumference, in. 39.3 ± 3.1 42.0 ± 3.5 34.6 ± 3.9
Albumin, g/dL 3.4 ± 0.3 3.2 ± 0.3 4.0 ± 0.2
Total protein, g/dL 5.9 ± 0.4 6.0 ± 0.4 6.6 ± 0.6

Data are presented as mean ± SD or number (percentage), as appropriate.

BMI , body mass index.

Bastian et al. Pregnancy decreases exposure to SL BUP. Am J Obstet Gynecol 2017 .


The PK parameter estimates are summarized in Table 2 . During pregnancy (average of PK-2 plus PK-3), the dose-normalized (plasma concentration/dose) area under the BUP plasma concentration-time curves (AUC 0→12 ) and Cmax BUP concentrations, as well as the BUP concentrations at 0 and 12 hours were significantly lower than during the postpartum period. None of these parameters differed significantly during pregnancy (ie, PK-2 vs PK-3). The Tmax BUP concentrations did not differ significantly between groups.



Table 2

Buprenorphine pharmacokinetic parameters





















































Parameter (untransformed raw data, mean ± SD) PK-2 (second trimester) N = 7 PK-3 (third trimester) N = 11 PK-P (Postpartum) N = 10 P value i (PK-2:PK-3) P value ii (½PK-2 plus ½PK-3:PK-P)
Dose, mg 8.0 ± 3.1 10.0 ± 3.7 8.0 ± 0.0 .3333 .0276 a
Tmax, h 1.6 ± 2.8 1.0 ± 1.1 1.9 ± 3.6 .9661 .8411
Dose-normalized Cmax, ng/mL 0.5 ± 0.1 0.5 ± 0.2 0.7 ± 0.3 .9964 .0365 a
Dose-normalized C0, ng/mL 0.1 ± 0.0 0.2 ± 0.2 0.3 ± 0.3 .8959 .0032 a
Dose-normalized C12, ng/mL 0.1 ± 0.0 0.1 ± 0.1 0.3 ± 0.3 .1260 .0001 a
Dose-normalized AUC, ng/h per milliliter 1.9 ± 1.4 2.2 ± 1.2 4.0 ± 2.5 .7953 .0068 a

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Apr 24, 2017 | Posted by in GYNECOLOGY | Comments Off on Dose-adjusted plasma concentrations of sublingual buprenorphine are lower during than after pregnancy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access