Skip to main content
Drug Metabolism and Disposition logoLink to Drug Metabolism and Disposition
. 2024 Aug;52(8):785–796. doi: 10.1124/dmd.124.001643

Development and Verification of a Full Physiologically Based Pharmacokinetic Model for Sublingual Buprenorphine in Healthy Adult Volunteers that Accounts for Nonlinear Bioavailability

Matthijs W van Hoogdalem 1, Ryota Tanaka 1, Trevor N Johnson 1, Alexander A Vinks 1, Tomoyuki Mizuno 1,
PMCID: PMC11257693  PMID: 38769016

Abstract

Sublingual buprenorphine is used for opioid use disorder and neonatal opioid withdrawal syndrome. The study aimed to develop a full physiologically based pharmacokinetic (PBPK) model that can adequately describe dose- and formulation-dependent bioavailability of buprenorphine. Simcyp (v21.0) was used for model construction. Four linear regression models (i.e., untransformed or log transformed for dose or proportion sublingually absorbed) were explored to describe sublingual absorption of buprenorphine across dose. Published clinical trial data not used in model development were used for verification. The PBPK model’s predictive performance was deemed adequate if the geometric means of ratios between predicted and observed (P/O) area under the curve (AUC), peak concentration (Cmax), and time to reach Cmax (Tmax) fell within the 1.25-fold prediction error range. Sublingual buprenorphine absorption was best described by a regression model with logarithmically transformed dose. By integrating this nonlinear absorption profile, the PBPK model adequately predicted buprenorphine pharmacokinetics (PK) following administration of sublingual tablets and solution across a dose range of 2–32 mg, with geometric mean (95% confidence interval) P/O ratios for AUC and Cmax equaling 0.99 (0.86–1.12) and 1.24 (1.09–1.40), respectively, and median (5th to 95th percentile) for Tmax equaling 1.11 (0.69–1.57). A verified PBPK model was developed that adequately predicts dose- and formulation-dependent buprenorphine PK following sublingual administration.

SIGNIFICANCE STATEMENT

The physiologically based pharmacokinetic (PBPK) model developed in this study is the first to adequately predict dose- and formulation-dependent sublingual buprenorphine pharmacokinetics. Accurate prediction was facilitated by the incorporation of a novel nonlinear absorption model. The developed model will serve as the foundation for maternal-fetal PBPK modeling to predict maternal and fetal buprenorphine exposures to optimize buprenorphine treatment for neonatal opioid withdrawal syndrome.

Introduction

Buprenorphine, administered as a sublingual tablet or solution, is used in the management of opioid use disorder (OUD). Buprenorphine acts as a partial agonist at the μ opioid receptor (Martin et al., 1976), as an antagonist at δ and κ opioid receptors (Leander 1988; Negus et al., 2002), and as a full agonist at the nociceptin/orphanin FQ opioid receptor (Wnendt et al., 1999). This intricate pharmacological profile gives rise to buprenorphine’s more desirable clinical properties compared with other opioids, such as lower abuse potential and reduced likelihood of fatal respiratory depression (Pergolizzi et al., 2010). Among Medicaid enrollees diagnosed with OUD, the use of buprenorphine increased from 28.1% to 37.3% between 2014 and 2018, making it the most prescribed medication to treat OUD (Donohue et al., 2021).

Opioid use during pregnancy is not uncommon. In 2019, 6.6% of pregnant women self-reported use of prescription opioids, of which 21.2% disclosed opioid misuse (Ko et al., 2020). Newborns prenatally exposed to opioids are at risk of developing neonatal opioid withdrawal syndrome (NOWS) after birth. NOWS is characterized by gastrointestinal dysfunction and neurologic excitability (Patrick et al., 2020) and requires pharmacological treatment in those neonates whose symptoms are otherwise insufficiently controlled (Wachman et al., 2018). Sublingually administered buprenorphine is an emerging treatment of NOWS (Simon et al., 2021), but current dosing strategies have been empirically established and lack a robust pharmacokinetic (PK) and pharmacodynamic (PD) rationale (Hjelmström et al., 2020). Neonatal buprenorphine PK is highly variable (Ng et al., 2015; Mizuno et al., 2021; van Hoogdalem et al., 2021), and recent physiologically based pharmacokinetic (PBPK) modeling and simulation by our group indicated that variability is likely driven by differences in the extent of sublingual absorption, biliary clearance, and cytochrome P450 (CYP) 3A4 activity, especially early in life (van Hoogdalem et al., 2022a). Strategies to improve the treatment of NOWS with buprenorphine include further improving our understanding of the complex PK/PD relationship and subsequently adjusting the starting dose to the expected PK profile of the neonate. Additionally, initial dosing could be tailored to the anticipated NOWS severity.

The severity of NOWS differs greatly between affected neonates, but symptoms tend to be more severe in newborns born at term (Liu et al., 2010), whose mothers used tobacco during pregnancy (Choo et al., 2004; Bakstad et al., 2009), and those who had greater opioid exposure in utero (van Hoogdalem et al., 2022b). Estimating the extent of prenatal opioid exposure is challenging. Intuitively, fetal opioid exposure may strongly correlate with maternal intake, but studies have failed to demonstrate a consistent relationship between maternal OUD medication dose and postnatal NOWS severity (Dashe et al., 2002; Bakstad et al., 2009; Dryden et al., 2009; Jones et al., 2014). This may be, in part, explained by the everchanging nature of maternal opioid PK during pregnancy and the likelihood that fetuses are more vulnerable to opioid effects at certain points during gestation (van Hoogdalem et al., 2022b). Maternal-fetal PBPK modeling offers a comprehensive framework that can incorporate the kaleidoscopic interplay of maternal and fetal factors that ultimately dictate prenatal opioid exposure (van Hoogdalem et al., 2022b). This, in turn, can open the way for precision treatment of NOWS based on the prenatally modeled severity.

Accurately predicting buprenorphine PK following sublingual administration is challenging since bioavailability is dependent on the formulation (tablet versus solution) (Nath et al., 1999, Schuh and Johanson 1999; Harris et al., 2004; Strain et al., 2004; Chawarski et al., 2005) and decreases with dose (Harris et al., 2004; Ciraulo et al., 2006; Dong et al., 2019). Several PBPK models for sublingual buprenorphine have been developed to date, but none have adequately integrated nonlinear bioavailability. Kalluri et al. (2017) constructed a full PBPK model, which was later expanded to a pregnancy PBPK model (Zhang et al., 2018), but others were not able to recreate these models due to the ambiguous description of sublingual absorption (Silva et al., 2022). Our group developed a neonatal minimal PBPK model (van Hoogdalem et al., 2022a), which was based on a model developed by Johnson et al. (2016), but given the neonatal application, the model was only verified for low doses and does not accurately capture reduced bioavailability with higher doses. In line with PBPK modeling best practices, it is recommended to develop and validate the model in healthy adults before extrapolating it to special populations. Therefore, to lay a strong foundation for planned maternal-fetal PBPK modeling, the aim of the present study was to develop a full PBPK model for buprenorphine that can adequately describe dose- and formulation-dependent bioavailability following sublingual administration.

Materials and Methods

PBPK Model Development

A full PBPK model for buprenorphine was constructed and verified using Simcyp (v21.0; Simcyp Limited, Sheffield, UK). A schematic representation of the PBPK model is shown in Fig. 1. Drug physiochemical and physiologic parameters used to build the PBPK model are shown in Table 1 (Bullingham et al., 1980; Holland et al., 1989; Avdeef et al., 1996; Kuhlman et al., 1996; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/20-733_Subutex_BioPharmr.pdf; Takahashi et al., 2001; Elkader and Sproule 2005; Picard et al., 2005; Chang and Moody 2009; Cubitt et al., 2009; Hassan et al., 2009; Moore et al., 2018; https://pubchem.ncbi.nlm.nih.gov/compound/644073). The fraction unbound in blood plasma was set to be 0.04 as reported by Elkader and Sproule (2005). The fraction absorbed was assumed to be 1. Concentration-time profiles reported by Dong et al. (2019), who administered buprenorphine sublingual tablets at doses of 2, 4, 8, 12, 16, and 24 mg to 82 opioid-naïve volunteers, were digitized using WebPlotDigitizer (v4.5, Ankit Rohatgi, Pacifica, CA). The first-order absorption rate constant from the gastrointestinal tract was optimized to accurately capture the peak concentration (Cmax) using the concentration-time profile following administration of a 24-mg buprenorphine sublingual tablet reported by Dong et al. (2019). A lag time parameter was integrated into the buprenorphine population PK model developed by Moore et al. (2018), and utilizing Bayesian estimation in MWPharm++ (v2.0.4; Mediware Incorporated, Prague, Czech Republic), the population PK model was fitted to the observed concentration-time profiles reported by Dong et al. (2019) for sublingual tablet doses of 2, 4, 8, 12, 16, and 24 mg to determine mean lag time. Elimination was based on in vitro enzyme kinetics data (Michaelis-Menten constant and Vmax) for CYP2C8-, CYP3A4-, UDP-glucuronosyltransferase (UGT)-1A1-, UGT1A3-, UGT2B7-, and UGT2B17-mediated metabolism, renal clearance, and biliary clearance. Renal clearance was calculated based on a mass balance study where 1% was excreted unchanged in urine (https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/20-733_Subutex_BioPharmr.pdf), with a total plasma clearance (CL) of 54.1 L/h (Bullingham et al., 1980).

TABLE 1.

Input data for the full PBPK model for buprenorphine

Parameter Value Reference
Physiochemical
Molecular weight (g/mol) 467.6 https://pubchem.ncbi.nlm.nih.gov/compound/644073
LogP 4.98 Avdeef et al., 1996
Compound type Ampholyte Avdeef et al., 1996
pKa (acid; phenol) 9.62 Avdeef et al., 1996
pKa (base; amine) 8.31 Avdeef et al., 1996
Blood binding
B/P 1 Bullingham et al., 1980
f u, plasma 0.04 Elkader and Sproule, 2005
Plasma binding components AGP Takahashi et al., 2001
Gastrointestinal tract absorption (first-order model)
fa 1a
ka (h−1) 0.016b
Lag time (h) 0.22c
f u, gut 0.4b
Qgut (L/h) 16.8d
Peff, man (10−4 cm/s) 6.83d
Caco-2 7.4:7.4 (10−6 cm/s) 66.7 Hassan et al., 2009
Sublinguale absorption (first-order model)
fa 1a
ka (h−1) 1b
Proportion of dose inhalede (tablet) (%) 38.1–19.7 × log(Dose)f
Proportion of dose inhalede (solution) (%) 53.3–25.6 × log(Dose)f
Distribution (full PBPK model)
Tissue-to-plasma partition coefficients (Kp)
Adiposeg 17.800 Takahashi et al., 2001
Boneh 1.603 Takahashi et al., 2001
Brainh 19.206 Takahashi et al., 2001
Guti 2.252 Takahashi et al., 2001
Hearth 1.714 Takahashi et al., 2001
Kidneyi 6.372 Takahashi et al., 2001
Liveri 8.695 Takahashi et al., 2001
Lungh 3.921 Takahashi et al., 2001
Muscleh 0.905 Takahashi et al., 2001
Pancreash 3.016 Takahashi et al., 2001
Skin 3.500 Holland et al., 1989
Spleenh 2.286 Takahashi et al., 2001
Predicted Vss (L/kg) 6.23d
Observed Vss (L/kg) 4.95 Kuhlman et al., 1996
Elimination
CYP2C8
Vmax (pmol/min per mg protein) 176.3 Picard et al., 2005
Km (μM) 12.4 Picard et al., 2005
CYP3A4
Vmax (pmol/min per mg protein) 520 Picard et al., 2005
Km (μM) 13.6 Picard et al., 2005
UGT1A1
Vmax (pmol/min per mg protein) 2870 Chang and Moody, 2009
Km (μM) 66.4 Chang and Moody, 2009
UGT1A3
Vmax (pmol/min per mg protein) 286 Chang and Moody, 2009
Km (μM) 202 Chang and Moody, 2009
UGT2B7
Vmax (pmol/min per mg protein) 173 Chang and Moody, 2009
Km (μM) 13.8 Chang and Moody, 2009
UGT2B17
Vmax (pmol/min per mg protein) 172 Chang and Moody, 2009
Km (μM) 9.6 Chang and Moody, 2009
f u, mic 0.1 Cubitt et al., 2009
CLrenal (L/h) 0.54j
CLbiliary (μl/min per million cells) 51 Johnson et al., 2016

AGP, α1-acid glycoprotein; B/P, blood-to-plasma ratio; CLbiliary, biliary clearance; CLrenal, renal clearance; fa, fraction absorbed; fu, gut, fraction unbound in enterocytes; fu, mic, fraction unbound in in vitro microsomal incubation; fu, plasma, fraction unbound in blood plasma; ka, first-order absorption rate constant; Km, Michaelis-Menten constant; Peff, man, human jejunum effective permeability; Qgut, nominal flow in gut model; UGT, UDP-glucuronosyltransferase; Vss, volume of distribution at steady-state.

aAssumed value.

bEmperically optimized to accurately capture the Cmax observed following administration of a 24-mg buprenorphine sublingual tablet as reported by Dong et al. (2019).

cDetermined through Bayesian estimation, employing concentration-time profiles reported by Dong et al. (2019) for sublingual tablet doses of 2, 4, 8, 12, 16, and 24 mg. A lag time (Tlag) parameter was integrated into the buprenorphine population PK model developed by Moore et al. (2018), and utilizing Bayesian estimation in MWPharm++, the population PK model was fitted to the observed concentration-time profiles at each dose level. Value represents the arithmetic mean of Bayesian-estimated Tlag across the 2–24-mg dose range.

dSimcyp predicted value.

eThe sublingual route of administration is not available in Simcyp; sublingual absorption is therefore mimicked by employing the first-order inhalation model in combination with the inhaled route of administration.

fDose is in mg, and logarithm base is 10. The value is calculated manually, and the computed proportion is then entered into the first-order inhalation model. Note that a coefficient of variation of 33.9% is applied to the administered dose to reflect variability in bioavailability; more details are provided in this manuscript.

gReported radioactivity at 24 h postinjection was used for calculation.

hReported radioactivity at 8 h postinjection was used for calculation.

iReported radioactivity at 1 h postinjection was used for calculation.

jCalculated by Johnson et al. (2016) based on a mass balance study where 1% was excreted unchanged in urine (https://www.accessdata.fda.gov/drugsatfda_docs/nda/2002/20-733_Subutex_BioPharmr.pdf), with a total plasma clearance of 54.1 L/h (Bullingham et al., 1980).

Fig. 1.

Fig. 1.

Full PBPK model structure. The sublingual route of administration is not available in Simcyp; sublingual absorption is therefore mimicked by employing the first-order inhalation model in combination with the inhaled route of administration. The proportion of the dose inhaled equals the proportion sublingually absorbed. The remaining fraction of the dose is swallowed.

The present model was based on a minimal PBPK model for buprenorphine developed earlier by our group (van Hoogdalem et al., 2022a), which, in turn, was adapted from a model described by Johnson et al. (2016). The minimal PBPK model was expanded to a full PBPK model by incorporating tissue-to-plasma partition coefficients (Kp). There were no reports of Kp values for buprenorphine from human tissue data. Therefore, Kp values were estimated using tissue distribution data in rats generally measured between 1 and 144 hours following subcutaneous injection of radiolabeled buprenorphine (Holland et al., 1989; Takahashi et al., 2001). Moment-dependent distribution of buprenorphine and its metabolites was considered when determining optimal time points to calculate Kp values; e.g., Kp values for gut, kidney, and liver were obtained using distribution data measured at 1 hour postdose to minimize measuring the distribution of buprenorphine metabolites rather than buprenorphine. The volume of distribution at steady state (Vss) was predicted in Simcyp.

As the sublingual route of administration is not available in Simcyp, sublingual absorption was simulated by employing two separate absorption models: one for lung and another for gastrointestinal absorption as described previously (van Hoogdalem et al., 2022a). In this approach, the proportion of the dose inhaled to undergo lung absorption mirrors the proportion that would be absorbed sublingually and circumvents first-pass metabolism. The remaining fraction is swallowed and is subject to the first-pass effect. Sublingual and gastrointestinal absorption was simulated based on the parameters summarized in Table 1.

Linear Regression Modeling of Sublingual Absorption

Plasma concentration-time data were extracted from dose-escalation (Schuh and Johanson 1999, Harris et al., 2004; Dong et al., 2019) studies (training data) using WebPlotDigitizer. Area under the curve [AUC; i.e., area under the curve from zero to infinity (AUC0–∞) and area under the curve in one dosing interval (AUC0τ) for single and multiple dose studies, respectively] and Cmax following sublingual tablet or solution administration were determined by fitting the buprenorphine population PK model reported by Moore et al. (2018) to these extracted concentration-time profiles using MWPharm++. All PK parameters were fitted with 6–20 concentration-time points for each trial. Subsequently, through multiple iterations with the PBPK model, the proportion of the dose designated for sublingual absorption in the PBPK model was determined to precisely recover the AUC and Cmax observed for doses between 2 and 32 mg administered in selected dose-escalation studies (Schuh and Johanson 1999; Harris et al., 2004; Dong et al., 2019), termed as the ideal proportion. Ideal proportion determination involved continuously adjusting the sublingual absorption proportion empirically during each run until the PBPK model predicted a geometric mean AUC and Cmax identical to that observed. Ideal proportions were determined separately for AUC and Cmax as different sublingual absorption proportions were often necessary to accurately replicate each exposure metric. The relationship between AUC- and Cmax-optimized ideal proportion and dose was explored for sublingual tablets and solution separately through linear regression modeling using the stats package (v4.1.2, R Core Team) for R (v4.1.2; R Foundation for Statistical Computing, Vienna, Austria). The following bivariate linear model was used (eq. 1):

graphic file with name dmd.124.001643_eq1.jpg

where Proportioni is the AUC- or Cmax-optimized ideal proportion (%) for clinical study i, α is the intercept, β is the slope, and Dose is the sublingual tablet or solution dose in milligrams. Visual inspection of the data indicated a linear or inverse exponential relationship between ideal proportion and dose. Therefore, four varieties of the linear model were explored, i.e., either Dose and Proportioni untransformed, or with Dose, Proportioni, or both logarithmically transformed using a decimal logarithm of base 10. Thus, in total, 16 linear regression analyses were performed, namely, four linear model varieties explaining four individual relationships (i.e., AUC- and Cmax-optimized ideal proportions versus sublingual tablet and solution doses, assessed as separate formulations). The linear model achieving the highest mean coefficient of determination across the four individual relationships was selected (Supplemental Table 1). AUC- and Cmax-optimized linear models were subsequently averaged to capture both AUC and Cmax using one equation, thereby obtaining two final linear models (one for sublingual tablets and one for sublingual solution) describing the relationship between ideal proportion and dose.

PBPK Model Verification and Evaluation

Following an extensive literature search for plasma buprenorphine PK data in healthy volunteers, the PBPK model’s predictive performance was assessed for intravenous and sublingual administration successively by determining the average-fold error (AFE) and absolute average-fold error (AAFE) between predicted and observed concentrations and the ratio between predicted and observed (P/O) AUC, CL, Cmax, and, in case of sublingual administration, time to reach Cmax (Tmax). The following equations were used to calculate AFE and AAFE (eqs. 2 and 3, respectively):

graphic file with name dmd.124.001643_eq2.jpg
graphic file with name dmd.124.001643_eq3.jpg

All data used for model verification were independent (test data), i.e., not used in the development of the PBPK or sublingual absorption model.

Predicted PK parameters were obtained by running virtual trials in Simcyp and represented the geometric mean of the virtual trial’s population. Virtual trials were conducted using the Sim-Healthy Volunteers population file. The population’s age (preferably age range, but mean age if no range was reported), proportion of females (50% was assumed for studies that did not report the participants’ sex), and administered buprenorphine dose and formulation were matched to that in the clinical study. All virtual trials involving sublingual administration of buprenorphine were conducted under the fasted state. For virtual trials in which buprenorphine was sublingually administered, a coefficient of variation of 33.9% was applied to the administered dose to reflect variability in bioavailability, which is consistent with the average variation in bioavailability after sublingual administration observed by Bullingham et al. (1982). The virtual cohort consisted of 100 individuals (10 individuals × 10 trials) for each simulation. The virtual trial duration was set to the time associated with the last reported observable concentration in the clinical study. For the sampling plan, predefined uniform intervals were selected, and the number of time samples was set to 16,000.

For clinical studies in which buprenorphine was intravenously administered, observed PK parameters were defined as those reported in the trial; missing values were calculated through noncompartmental analysis using Edsim++ (v2.0.4; Mediware Incorporated, Prague, Czech Republic). Clinical studies rarely determined a true Cmax following intravenous administration. Instead, Cmax generally represented the first concentration measured a few minutes after completion of a bolus injection. Therefore, to match predicted and observed Cmax, predicted Cmax was defined as the modeled concentration at the time point associated with the first concentration measured.

For clinical studies in which buprenorphine was sublingually administered, observed PK parameters were, similar as described under Linear Regression Modeling of Sublingual Absorption, obtained through Bayesian estimation by fitting the buprenorphine population PK model reported by Moore et al. (2018) to concentration-time data extracted from publications using WebPlotDigitizer. Reported PK parameter values were not used as some studies employed sparse sampling strategies, which limited the robustness of time-associated (i.e., Tmax and Cmax) and exposure-dictated (i.e., AUC) PK parameters. In the interest of consistency, all concentration-time profiles of sublingually administered buprenorphine for each clinical study were digitized and used to estimate PK parameters through Bayesian estimation.

Potential bias in the PBPK model’s prediction following sublingual administration was evaluated using predicted-versus-observed AUC, Cmax, and Tmax and dose-versus-respective-P/O-ratio plots.

Statistical Analysis

Geometric means and 95% confidence intervals (CIs) of PK parameter P/O ratios were calculated using the DescTools package (v0.99.44; Signorell et mult. al.) for R. Normal distribution of P/O ratios was examined through the Shapiro-Wilk test. The predictive performance of the PBPK model was deemed adequate if the geometric means of PK parameter P/O ratios fell between 0.8-fold and 1.25-fold (1.25-fold prediction error range). In addition to assessing whether geometric mean PK parameter P/O ratio fell within the relatively narrow 1.25-fold prediction error range, the proportion of all AAF, AAFE, and PK parameter P/O ratios falling within the wider twofold prediction error range was determined.

Results

Verification of the PBPK Model’s Predictive Performance following Intravenous Administration

The structure of the full PBPK model was first externally verified by determining the AFE, AAFE, and P/O ratios of AUC0–∞, CL, and Cmax following intravenous administration of buprenorphine in healthy volunteers. Twelve PK studies, spanning a dose range of 0.3–16 mg and including a total of 69 subjects (aged 20–66.8 years) with 89 concentration-time profiles, were used for intravenous model verification (Table 2) (Bullingham et al., 1982; Kuhlman et al., 1996; Mendelson et al., 1997; Huestis et al., 2013; Bai et al., 2016; Lim et al., 2019). For all 12 PK studies, the AFE and AAFE, as well as the P/O ratios of AUC0–∞, CL, and Cmax, fell within the twofold prediction error range. Geometric mean (95% CI) AUC0–∞, CL, and Cmax P/O ratios were 1.01 (0.90–1.13), 0.95 (0.84–1.08), and 0.91 (0.78–1.05), respectively, indicating adequate predictive performance of these PK parameters following intravenous administration across a wide dose range in healthy volunteers. All predicted-versus-observed buprenorphine concentration-time profiles following intravenous administration are shown in Fig. 2. The relative contribution of individual metabolic and elimination pathways involved in the clearance of buprenorphine is shown in Supplemental Fig. 1, which was comparable to the minimal PBPK model reported previously (van Hoogdalem et al., 2022a).

TABLE 2.

Predicted and observed pharmacokinetic parameters of buprenorphine following intravenous administration

Clinical Trial Dose (mg) Route of Administration n Female (%) Mean Age [range] (years) AFE AAFE AUC0–∞ (ng × h/mL) CL (L/h) Cmax
(ng/mL)
Bullingham et al., 1982 0.3 Intravenous (1 min) 5 60 66.8 0.66 1.71 Predicted Observed 5.96 5.80a 50.3 51.8b 1.390.96
P/O ratio 1.03 0.97 1.45
Bullingham et al., 1982 0.3 Intravenous (1 min) 5 60 64.2 1.34 1.39 PredictedObserved 4.763.14a 63.095.7b 1.391.08
P/O ratio 1.52 0.66 1.29
Bullingham et al., 1982 0.3 Intravenous (1 min) 5 60 66.0 1.09 1.62 PredictedObserved 4.653.20a 64.493.8b 1.380.95
P/O ratio 1.45 0.69 1.45
Bai et al., 2016 0.3 Intravenous (2 min) 24 24.0 35.5 (20–53) 0.59 1.72 PredictedObserved 4.805.20 62.557.7b 1.712.32
P/O ratio 0.92 1.08 0.74
Lim et al., 2019 0.3 Intravenous (5 min) 14 NR 25 0.87 1.36 PredictedObserved 4.534.09 66.277.7 1.932.73
P/O ratio 1.11 0.85 0.71
Mendelson et al., 1997 1 Intravenous (30 min) 6 16.7 29 (21–38) 1.12 1.31 PredictedObserved 15.818.4 63.262.5 13.114.3
P/O ratio 0.86 1.01 0.92
Kuhlman et al., 1996 1.2 Intravenous (1 min) 5 0.0 34.4 (27–40) 0.85 1.58 PredictedObserved 19.717.4 60.876.8 25.437.5
P/O ratio 1.13 0.79 0.68
Huestis et al., 2013 2 Intravenous (1 min) 5 0.0 34.6 (32–39) 0.79 1.49 Predicted Observed 33.341.4 60.149.8 15.719.3
P/O ratio 0.80 1.21 0.81
Huestis et al., 2013 4 Intravenous (1 min) 5 0.0 34.6 (32–39) 0.73 1.59 PredictedObserved 66.675.9 60.153.2 31.444.0
P/O ratio 0.88 1.13 0.71
Huestis et al., 2013 8 Intravenous (1 min) 5 0.0 34.6 (32–39) 0.74 1.54 PredictedObserved 133.2153.3 60.152.4 62.985.7
P/O ratio 0.87 1.15 0.73
Huestis et al., 2013 12 Intravenous (1 min) 5 0.0 34.6 (32–39) 0.73 1.46 PredictedObserved 199.8245.1 60.154.7 94.3107.9
P/O ratio 0.82 1.10 0.87
Huestis et al., 2013 16 Intravenous (1 min) 5 0.0 34.6 (32–39) 0.85 1.43 Predicted Observed 266.4269.1 60.160.0 125.8134.0
P/O ratio 0.99 1.00 0.94
Geo. meanc 1.01 0.95 0.91
(95% CI) (0.90–1.13) (0.84–1.08) (0.78–1.05)

NR, not reported.

aCalculated through noncompartmental analysis.

bCalculated following CL = Dose/AUC0–∞.

cGeometric mean of P/O ratios.

Fig. 2.

Fig. 2.

PBPK model–predicted and observed concentration-time profiles of buprenorphine following intravenous administration. Blue solid line and shaded area represent the mean and 5th to 95th percentile range of the virtual population (n = 100), respectively. Open circles represent individual observations. Closed circles and whiskers represent mean and standard deviation of the observations, respectively. References for reported observations are provided in Table 2.

Integrating Nonlinear Sublingual Absorption into the PBPK Model

Linear regression models with logarithmically transformed dose best described the relationships between AUC- and Cmax-optimized ideal proportions and sublingual tablet and solution doses (mean coefficient of determination, 0.756; Fig. 3 and Supplemental Table 1), which indicated that sublingual buprenorphine absorption is nonlinear across dose. By averaging the AUC- and Cmax-optimized linear regression models, two final absorption equations were obtained, namely, proportion of the dose sublingually absorbed equals 38.1 – 19.7 × log(Dose) and 53.3 – 25.6 × log (Dose) for sublingual tablets and solution, respectively. These equations were integrated into the PBPK model as shown in Fig. 1.

Fig. 3.

Fig. 3.

Proportion of the dose required by the physiologically-based pharmacokinetic (PBPK) model to be sublingually absorbed to exactly recover the (A) AUC (i.e., AUC0–∞ and AUC0–τ for single and multiple dose studies, respectively) and (B) Cmax observed in the clinical trial (i.e., ideal proportion) across dose. Blue and orange circles, triangles, and diamonds represent sublingual tablet and solution data obtained from Harris et al. (2004), Schuh and Johanson (1999), and Dong et al. (2019), respectively. Blue and orange dotted lines represent linear regression models with logarithmically transformed dose for buprenorphine tablets and solution, respectively. Respective shaded areas represent the 95% CI of the regression models. Associated linear-log equations are shown in the upper right corners (where dose is in milligrams and logarithm base is 10), with coefficients of determination (R2) shown in the lower-left corners. The final buprenorphine PBPK model uses the average of the AUC- and Cmax-optimized equations, i.e., proportion sublingually absorbed equals 38.1–19.7 × log(Dose) and 53.3 – 25.6 × log(Dose) for sublingual tablets and solution, respectively.

Verification and Evaluation of the PBPK Model’s Predictive Performance following Sublingual Administration

The PBPK model with the developed description of nonlinear sublingual buprenorphine absorption was subsequently externally verified by determining the AFE and AAFE, as well as the P/O ratios of AUC, Cmax, and Tmax following sublingual administration of buprenorphine tablets and solution separately. For verification of the PBPK model’s predictive performance following sublingually administered tablets, 16 PK studies, spanning a dose range of 2–32 mg and including a total of 296 subjects (aged 19–54) with 419 concentration-time profiles, were used (Table 3) (Nath et al., 1999; McAleer et al., 2003; Chawarski et al., 2005; Ciraulo et al., 2006; Moody et al., 2011; Jönsson et al., 2018). For all 16 PK studies, the AFE and AAFE, as well as the P/O ratios of AUC, Cmax, and Tmax, fell within the twofold prediction error range. Geometric mean (95% CI) AUC and Cmax P/O ratios were 0.96 (0.82–1.12) and 1.20 (1.05–1.37), respectively, and the median (5th to 95th percentile) for Tmax was 1.12 (0.69–1.53).

Table 3.

Predicted and observed buprenorphine pharmacokinetic parameters following administration of sublingual tablets

Observed PK parameters were obtained through Bayesian estimation by fitting the buprenorphine population PK model reported by Moore et al. (2018) to extracted concentration-time profiles.

Clinical Trial Dose (mg) Route of Administration n Female (%) Mean Age [range] (years) AFE AAFE AUCa (ng×h/mL) Cmax
(ng/mL)
Tmax (h)
McAleer et al., 2003 2 Sublingual, tablet 27 0.0 (19–42) 0.77 1.48 Predicted 7.32 1.37 1.14
Observed 10.3 1.47 1.48
P/O ratio 0.71 0.93 0.77
Ciraulo et al., 2006 4 Sublingual, tablet 23 30.4 34.5 1.87 1.90 Predicted 15.9 2.67 1.12
Observed 9.62 1.87 1.00
P/O ratio 1.65 1.43 1.12
Jönsson et al., 2018 4 Sublingual, tablet 61 41.0 31.4 (19–54) 0.92 1.59 Predicted 13.7 2.31 1.08
Observed 21.8 2.14 1.69
P/O ratio 0.63 1.08 0.64
Nath et al., 1999 8 Sublingual, tablet 6 0.0 28 (23–42) 1.11 1.52 Predicted 22.6 3.52 1.15
Observed 23.5 2.95 1.10
P/O ratio 0.96 1.19 1.05
McAleer et al., 2003 8 Sublingual, tablet 27 0.0 (19–42) 0.82 1.32 Predicted 22.9 3.49 1.14
Observed 29.1 3.84 1.27
P/O ratio 0.79 0.91 0.90
Ciraulo et al., 2006 8 Sublingual, tablet 23 30.4 34.5 1.42 1.57 Predicted 27.1 4.15 1.12
Observed 20.8 2.47 0.99
P/O ratio 1.30 1.68 1.13
McAleer et al., 2003 12 Sublingual, tablet 27 0.0 (19–40) 0.81 1.30 Predicted 31.0 4.35 1.15
Observed 41.0 4.81 1.12
P/O ratio 0.76 0.90 1.03
McAleer et al., 2003 16 Sublingual, tablet 27 0.0 (19–40) 0.77 1.33 Predicted 38.2 4.98 1.15
Observed 52.7 6.11 0.79
P/O ratio 0.72 0.82 1.46
Chawarski et al., 2005 16 Sublingual, tablet, m.d. 18 29.5 37.8 1.71 1.71 Predicted 45.5 6.68 1.12
Observed 31.2 3.45 0.71
P/O ratio 1.46 1.94 1.58
Ciraulo et al., 2006 16 Sublingual, tablet 23 30.4 34.5 1.26 1.40 Predicted 45.0 5.93 1.12
Observed 42.0 4.11 0.96
P/O ratio 1.07 1.44 1.17
Moody et al., 2011 16 Sublingual, tablet, m.d. 11 100.0 41.5 0.92 1.24 Predicted 46.8 6.87 1.00
Observed 57.8 6.58 0.90
P/O ratio 0.81 1.04 1.11
Moody et al., 2011 16 Sublingual, tablet, m.d. 20 0.0 35.7 1.27 1.30 Predicted 45.3 6.58 1.17
Observed 40.9 4.54 1.05
P/O ratio 1.11 1.45 1.11
Jönsson et al., 2018 16 Sublingual, tablet 64 40.6 32.1 (20–51) 1.00 1.52 Predicted 39.7 5.17 1.09
Observed 56.3 5.29 1.54
P/O ratio 0.71 0.98 0.71
Chawarski et al., 2005 24 Sublingual, tablet, m.d. 19 29.5 37.8 1.24 1.26 Predicted 60.1 8.01 1.13
Observed 56.8 6.86 0.91
P/O ratio 1.06 1.17 1.24
Ciraulo et al., 2006 24 Sublingual, tablet 23 30.4 34.5 1.18 1.38 Predicted 59.5 6.81 1.13
Observed 61.7 5.08 0.75
P/O ratio 0.96 1.34 1.51
Chawarski et al., 2005 32 Sublingual, tablet, m.d. 20 29.5 37.8 1.51 1.51 Predicted 72.2 8.76 1.13
Observed 55.5 6.17 0.97
P/O ratio 1.30 1.42 1.16
Geo. meanc 0.96 1.20 1.12c
(95% CI) (0.82–1.12) (1.05–1.37) (0.69–1.53)

m.d., multiple doses.

aAUC0–∞ and AUC0–τ for single and multiple dose studies, respectively.

bGeometric mean of P/O ratios.

cMedian (5th to 95th percentile) of P/O ratios.

For verification of the predictive performance following administration of sublingual solution, seven PK studies, spanning a dose range of 2–16 mg and including a total of 75 subjects (aged 21–42) with 81 concentration-time profiles, were used (Table 4) (Kuhlman et al., 1996; Mendelson et al., 1997; Nath et al., 1999; Chawarski et al., 2005). For all seven PK studies, the AFE and AAFE, as well as the P/O ratios of AUC and Tmax, fell within the twofold prediction error range, except for the AAFE of the 4-mg dose in the study by Kuhlman et al. (1996). The P/O ratio for Cmax fell within the twofold prediction error range in six out of seven (85.7%) PK studies. Geometric mean (95% CI) AUC and Cmax P/O ratios were 1.05 (0.75–1.46) and 1.34 (0.95–1.90), respectively, and the median (5th to 95th percentile) for Tmax was 1.02 (0.75–1.69). For three studies in which both intravenous and sublingual PK were reported, the P/O ratios of bioavailability were within the twofold prediction error range in all cases and 1.25-fold in two cases, and the geometric mean was also within the 1.25-fold range (Supplemental Table 2).

Table 4.

Predicted and observed buprenorphine pharmacokinetic parameters following administration of sublingual solution

Unless stated otherwise, observed PK parameters were obtained through Bayesian estimation by fitting the buprenorphine population PK model reported by Moore et al. (2018) to extracted concentration-time profiles.

Clinical Trial Dose (mg) Route of Administration n Female (%) Mean Age [range] (years) AFE AAFE AUCa (ng×h/mL) Cmax
(ng/mL)
Tmax (h)
Mendelson et al., 1997 2 Sublingual, solution (3 min hold) 6 16.7 29 (21–38) 1.59 1.78 Predicted 9.36 1.94 1.11
Observed 14.3b 1.60b 1.25b
P/O ratio 0.65 1.21 0.89
Mendelson et al., 1997 2 Sublingual, solution (5 min hold) 6 16.7 29 (21–38) 1.60 1.80 Predicted 9.36 1.94 1.11
Observed 13.2b 1.72b 1.62b
P/O ratio 0.71 1.13 0.69
Kuhlman et al., 1996 4 Sublingual, solution 6 0.0 34.4 (27–40) 0.79 2.57 Predicted 17.1 3.28 1.15
Observed 15.0 3.22 0.60
P/O ratio 1.14 1.02 1.92
Nath et al., 1999 8 Sublingual, solution 6 0.0 28 (23–42) 0.79 1.35 Predicted 28.9 5.19 1.14
Observed 34.6 6.72 1.02
P/O ratio 0.84 0.77 1.12
Chawarski et al., 2005 8 Sublingual, solution, 18 29.5 37.8 1.52 1.52 Predicted 35.0 6.38 1.12
Observed 25.4 3.19 1.18
m.d. P/O ratio 1.38 2.00 0.95
Chawarski et al., 2005 12 Sublingual, solution, 19 29.5 37.8 1.59 1.59 Predicted 47.3 8.27 1.12
Observed 33.7 4.50 0.98
m.d. P/O ratio 1.40 1.84 1.14
Chawarski et al., 2005 16 Sublingual, solution, 20 29.5 37.8 1.83 1.83 Predicted 58.1 9.80 1.12
Observed 36.4 4.91 1.10
m.d. P/O ratio 1.60 2.00 1.02
Geo. meanc 1.05 1.34 1.02d
(95% CI) (0.75–1.46) (0.95–1.90) (0.75–1.69)

m.d., multiple doses.

aAUC0–∞ and AUC0–τ for single and multiple dose studies, respectively.

bValue as reported in the original study, i.e., not obtained through Bayesian estimation.

cGeometric mean of P/O ratios.

dMedian (5th to 95th percentile) of P/O ratios.

On average for tablet and solution formulations, the geometric mean (95% CI) AUC and Cmax P/O ratios were 0.99 (0.86–1.12) and 1.24 (1.09–1.40), respectively, and the median (5th to 95th percentile) for Tmax was 1.11 (0.69–1.57). All predicted-versus-observed buprenorphine concentration-time profiles following sublingual administration are shown in Fig. 4. Predicted-versus-observed plots for AUC and Tmax did not reveal a bias as data points were symmetrically distributed across the line of equality (Fig. 5). Similarly, dose-versus-P/O-ratio plots suggested an unbiased prediction of AUC and Tmax across dose (Fig. 6), although clinical studies in which participants received sublingual solution were relatively few, and the dose range was smaller. Although comparison plots indicated a modest trend toward overpredicting Cmax, especially for high doses, the PBPK model’s predictive performance of buprenorphine PK following sublingual administration seemed to overall be adequate for both formulations across a wide dose range in healthy volunteers.

Fig. 4.

Fig. 4.

PBPK model–predicted and observed concentration-time profiles of buprenorphine following sublingual (s.l.) administration. Blue solid line and shaded area represent the mean and 5th to 95th percentile range of the virtual population (n = 100), respectively. Open circles represent individual observations. Closed circles and whiskers represent mean and standard deviation of the observations, respectively. References for reported observations are provided in Tables 3 and 4. m.d., multiple doses.

Fig. 5.

Fig. 5.

Comparison plots between predicted and observed for the final sublingual buprenorphine PBPK model, showing PBPK model–based predicted versus observed (A) AUC (i.e., AUC0–∞ and AUC0–τ for single and multiple dose studies, respectively), (B) Cmax, and (C) Tmax. In each panel, the solid black line represents the line of equality, where grayscale dashed, dot-and-dash, and dotted lines represent 1.25-, 1.5-, and twofold prediction error ranges, respectively. Curved blue solid lines represent locally estimated scatterplot smoothing curves.

Fig. 6.

Fig. 6.

Comparison plots between dose and P/O ratio for the final sublingual buprenorphine PBPK model, showing dose versus the P/O ratio (A) AUC (i.e., AUC0–∞ and AUC0–τ for single and multiple dose studies, respectively), (B) Cmax, and (C) Tmax as listed in Tables 3 and 4. Sublingual tablet and solution doses are represented by diamonds and circles, respectively. In each panel, the solid black line represents the line of equality, where descending shades of blue filled areas represent 1.25-, 1.5-, and twofold prediction error ranges, respectively. Curved black dashed lines represent locally estimated scatterplot smoothing curves.

Discussion

This is the first study to describe dose- and formulation-dependent sublingual buprenorphine absorption across a wide dose range through PBPK modeling. The developed model will serve as a foundation to build a maternal-fetal PBPK model for buprenorphine, which can be used to explore the relationship between fetal buprenorphine exposure and the severity of NOWS postnatally. By integrating a novel description of nonlinear sublingual buprenorphine absorption, the model adequately predicted PK following administration of sublingual tablets and solution. First, the full PBPK model structure was successfully externally verified using published intravenous PK data. Subsequently, a total of 23 published PK studies not used for model development, in which 371 healthy volunteers received buprenorphine as either sublingual tablet or solution across a dose range of 2–32 mg, were used to verify the final PBPK model. Geometric mean P/O ratios of AUC, Cmax, and Tmax were close to unity and fell within the 1.25-fold prediction error range. Comparison plots between dose and P/O ratio indicated unbiased prediction of all PK parameters, except for Cmax, which suggested a moderate trend toward overprediction, especially for high doses.

Previous studies have demonstrated nonlinear PK of sublingually administered buprenorphine (either as tablet or solution) across the entire dose range used for the management of OUD (Harris et al., 2004; Ciraulo et al., 2006; Dong et al., 2019). PK following intravenous administration, in contrast, is linear (Huestis et al., 2013), which strongly suggests that nonlinearity observed under sublingual dosing is driven by varying bioavailability rather than by changes in clearance. Various mechanisms have been proposed to explain nonlinear bioavailability, including varying dissolution degrees and times between tablet strengths (Harris et al., 2004), where high-dosed formulations may need to be kept in situ longer to allow maximal absorption, thereby increasing the risk of swallowing relatively more of the dose. In addition, buprenorphine sequesters in oral tissues (Cone et al., 1990), which decreases the concentration gradient that drives sublingual absorption of buprenorphine. The absorption model proposed in this study captures nonlinear bioavailability observed clinically. It is, however, important to note that the model was developed using PK data across a dose range of 2–32 mg (Schuh and Johanson, 1999; Harris et al., 2004; Dong et al., 2019). We caution against applying the absorption model outside this dose interval.

The developed PBPK model assumed the same lag time for both the tablet and solution. We investigated the lag time for the tablet and solution formulations by estimating it using concentration-time data from the 11 published PK studies. The results indicated no significant difference in estimated lag time between the formulations (tablet, 0.20 ± 0.09; solution, 0.17 ± 0.08; P = 0.37), indicating that using the same lag time for both tablet and solution is reasonable.

The developed PBPK model tended to underpredict the buprenorphine concentrations in the terminal elimination phase, where the discrepancy is particularly noticeable following intravenous administration (Fig. 2), which was consistent with the previous buprenorphine PBPK model reported by Kalluri et al. (2017). One possible explanation is the model's lack of consideration for enterohepatic recirculation, which is a significant limitation, particularly as buprenorphine is partially metabolized by UGTs, and its glucuronide conjugate can be reabsorbed after deconjugation by intestinal bacteria (McAleer et al., 2003). In addition, low observed concentrations might be influenced by assay variability as assay error is typically large near the lower limit of quantitation.

The developed model has a few limitations. First, Kp values used to describe the buprenorphine tissue distribution were obtained from rat data as there are no reports of buprenorphine Kp values for human tissues. More importantly, distribution in rats was not measured under strict steady-state conditions (Holland et al., 1989; Takahashi et al., 2001), which limits the robustness of the Kp values estimated in this study. Nevertheless, using these Kp values, observed concentrations were well captured by the PBPK model, and Vss was furthermore calculated at 6.23 L/kg in Simcyp, which approximates 4.95 L/kg observed clinically (Kuhlman et al., 1996). It is important to acknowledge that in this clinical study, Vss was derived from clearance multiplied by the mean residence time following an intravenous bolus injection, which may not accurately represent steady-state conditions, suggesting that the observed Vss could be an estimate rather than a precise measurement. We explored using the Rodgers and Rowland (2006) method as an alternative to predict tissue distribution (method 2 in Simcyp), but this resulted in an estimated Vss of 23.0 L/kg, which would necessitate the application of an empirically identified Kp scalar to recover the observed Vss. Instead, we deemed distribution estimated from rat data to be more in line with the physiologic rationale of PBPK modeling. Furthermore, Takahashi et al. (2001) reported degrees of in vitro plasma protein binding in rats and humans of 88.9%–95.3% and 94.4%–96.7%, respectively, suggesting no interspecies differences. The buprenorphine concentrations in tissues were not verified in the present model as there were no available concentration-time data in human tissues, other than placenta, following sublingual administration of buprenorphine. Further clinical study investigating the buprenorphine tissue distribution in humans is warranted to verify the prediction of tissue concentrations, although the extensive verification in this study indicated the adequate prediction of the plasma concentration.

Another limitation is that the present model overestimates Cmax modestly following sublingual administration of buprenorphine tablets and solution (geometric mean P/O ratios of 1.20 and 1.34, respectively). Manual parameter estimation of ideal proportion would preferably have yielded one and the same value to recover both observed AUC and Cmax simultaneously for each dose, but ideal proportion values for AUC and Cmax diverged, especially at the lower and upper limits of the dose spectrum (Fig. 3). This indicates an oversimplification of sublingual absorption in the current PBPK model. The model accounts for differences in the total transfer of buprenorphine across oral mucosa, but the rate of this process is likely variable across dose and formulation. Absorption rate differences were not integrated into the PBPK model, and AUC- and Cmax-optimized nonlinear absorption models were instead averaged, leading to a modest overestimation of Cmax overall. To understand the implication of this overestimation, it is worthwhile to briefly review the PK/PD relationship of buprenorphine and, specifically, the degree by which its PD effect is explained by Cmax compared with AUC. Yassen et al. (2007) characterized the PK/PD relationship of buprenorphine in healthy volunteers with respect to its respiratory depressant effect, which is an unambiguous marker for buprenorphine’s penetration into the central nervous system and its receptor association/dissociation kinetics at the μ opioid receptor (Boom et al., 2012). They estimated the time required for concentration at the effect site to reach 50% of the plasma concentration for buprenorphine at 75.3 minutes (Yassen et al., 2007), which, relative to other opioids, indicates a slow onset of action but a longer duration, where its effect is only marginally driven by Cmax (Upton et al., 1997). Since the developed PBPK model adequately predicts AUC following sublingual administration of buprenorphine, we believe the implications of modestly overestimating Cmax are therefore limited. It is important to note, though, that we are not discounting the significance of Cmax altogether, particularly as the primary determinants of buprenorphine-induced NOWS in the fetus remain unclear. Consequently, it is essential for the PBPK model to adequately predict all PK parameters and exposure metrics, enabling a thorough investigation into their association with NOWS severity.

In conclusion, the full PBPK model developed in this study is the first to adequately capture buprenorphine PK following sublingual administration (either as tablet or solution) across a wide dose range. The model provides valuable insights into the mechanisms that underly complex sublingual buprenorphine PK. Potential applications of the model include using it to optimize the treatment of OUD with buprenorphine, but for our group specifically, the model forms the basis for planned maternal-fetal PBPK modeling endeavors (van Hoogdalem et al., 2024). Improving the treatment of NOWS requires tailoring of pharmacotherapy based on the expected severity of withdrawal symptoms. Predicting fetal drug exposure prior to birth might better inform the severity of NOWS after birth than the maternal dose. Maternal-fetal PBPK modeling of buprenorphine facilitates estimation of prenatal buprenorphine exposure throughout gestation based on maternal intake, which opens the way for examining the likely link it has with postnatal withdrawal severity. This, in turn, could enable maternal-fetal PBPK model–informed precision dosing of buprenorphine, which is expected to improve the clinical outcomes of neonates affected by NOWS. The thoroughly verified PBPK model for buprenorphine developed in this study forms the fundament for this task.

Data Availability

The data presented in this study are available upon written and reasonable request from the corresponding authors.

Abbreviations

AAFE

absolute average-fold error

AFE

average-fold error

AUC

area under the curve

AUC0–τ

area under the curve in one dosing interval

AUC0–∞

area under the curve from zero to infinity

CI

confidence interval

CL

clearance

Cmax

peak concentration

CYP

cytochrome P450

Kp

tissue-to-plasma partition coefficients

NOWS

neonatal opioid withdrawal syndrome

OUD

opioid use disorder

P/O

predicted and observed

PBPK

physiologically based pharmacokinetic

PD

pharmacodynamic

PK

pharmacokinetic

Tmax

time to reach Cmax

UGT

UDP-glucuronosyltransferase

Vss

volume of distribution at steady state

Authorship Contributions

Participated in research design: van Hoogdalem, Vinks, Mizuno.

Conducted experiments: van Hoogdalem, Tanaka, Johnson, Mizuno.

Performed data analysis: van Hoogdalem, Tanaka, Johnson, Vinks, Mizuno.

Wrote or contributed to the writing of the manuscript: van Hoogdalem, Tanaka.

Footnotes

The work was supported in part by National Institutes of Health National Center for Advancing Translational Sciences [Grant 2UL1TR001425-05A1] (to T.M.) and the Maternal and Pediatric Precision in Therapeutics Knowledge & Research Coordination Center of the Eunice Kennedy Shriver National Institute of Child Health and Human Development [Grant 1P30HD106451] (to T.M.). M.W.v.H. was supported by the Rieveschl/Parke-Davis Doctoral Candidacy Scholarship of the University of Cincinnati. R.T. was supported by the Japan Research Foundation for Clinical Pharmacology. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health and the Eunice Kennedy Shriver National Institute of Child Health and Human Development.

M.W.v.H. is an employee of Johnson & Johnson. The company had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results. T.N.J. is an employee of Certara UK Limited, Simcyp Division. No other authors declared an actual or perceived conflict of interest with the contents of this article.

Inline graphicThis article has supplemental material available at dmd.aspetjournals.org.

References

  1. Avdeef A, Barrett DA, Shaw PN, Knaggs RD, Davis SS (1996) Octanol-, chloroform-, and propylene glycol dipelargonat-water partitioning of morphine-6-glucuronide and other related opiates. J Med Chem 39:4377–4381. [DOI] [PubMed] [Google Scholar]
  2. Bai SA, Xiang Q, Finn A (2016) Evaluation of the pharmacokinetics of single- and multiple-dose buprenorphine buccal film in healthy volunteers. Clin Ther 38:358–369. [DOI] [PubMed] [Google Scholar]
  3. Bakstad B, Sarfi M, Welle-Strand GK, Ravndal E (2009) Opioid maintenance treatment during pregnancy: occurrence and severity of neonatal abstinence syndrome. A national prospective study. Eur Addict Res 15:128–134. [DOI] [PubMed] [Google Scholar]
  4. Boom M, Niesters M, Sarton E, Aarts L, Smith TW, Dahan A (2012) Non-analgesic effects of opioids: opioid-induced respiratory depression. Curr Pharm Des 18:5994–6004. [DOI] [PubMed] [Google Scholar]
  5. Bullingham RE, McQuay HJ, Moore A, Bennett MR (1980) Buprenorphine kinetics. Clin Pharmacol Ther 28:667–672. [DOI] [PubMed] [Google Scholar]
  6. Bullingham RE, McQuay HJ, Porter EJ, Allen MC, Moore RA (1982) Sublingual buprenorphine used postoperatively: ten hour plasma drug concentration analysis. Br J Clin Pharmacol 13:665–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Chang Y, Moody DE (2009) Glucuronidation of buprenorphine and norbuprenorphine by human liver microsomes and UDP-glucuronosyltransferases. Drug Metab Lett 3:101–107. [DOI] [PubMed] [Google Scholar]
  8. Chawarski MC, Moody DE, Pakes J, O’Connor PG, Schottenfeld RS (2005) Buprenorphine tablet versus liquid: a clinical trial comparing plasma levels, efficacy, and symptoms. J Subst Abuse Treat 29:307–312. [DOI] [PubMed] [Google Scholar]
  9. Choo RE, Huestis MA, Schroeder JR, Shin AS, Jones HE (2004) Neonatal abstinence syndrome in methadone-exposed infants is altered by level of prenatal tobacco exposure. Drug Alcohol Depend 75:253–260. [DOI] [PubMed] [Google Scholar]
  10. Ciraulo DA, Hitzemann RJ, Somoza E, Knapp CM, Rotrosen J, Sarid-Segal O, Ciraulo AM, Greenblatt DJ, Chiang CN (2006) Pharmacokinetics and pharmacodynamics of multiple sublingual buprenorphine tablets in dose-escalation trials. J Clin Pharmacol 46:179–192. [DOI] [PubMed] [Google Scholar]
  11. Cone EJ, Dickerson SL, Darwin WD, Fudala P, Johnson RE (1990) Elevated drug saliva levels suggest a “depot-like” effect in subjects treated with sublingual buprenorphine. NIDA Res Monogr 105:569. [PubMed] [Google Scholar]
  12. Cubitt HE, Houston JB, Galetin A (2009) Relative importance of intestinal and hepatic glucuronidation-impact on the prediction of drug clearance. Pharm Res 26:1073–1083. [DOI] [PubMed] [Google Scholar]
  13. Dashe JS, Sheffield JS, Olscher DA, Todd SJ, Jackson GL, Wendel GD (2002) Relationship between maternal methadone dosage and neonatal withdrawal. Obstet Gynecol 100:1244–1249. [DOI] [PubMed] [Google Scholar]
  14. Dong RWang HLi DLang LGray FLiu YLaffont CMYoung MJiang JLiu Z, et al. (2019) Pharmacokinetics of sublingual buprenorphine tablets following single and multiple doses in chinese participants with and without opioid use disorder. Drugs R D 19:255–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Donohue JMJarlenski MPKim JYTang LAhrens KAllen LAustin ABarnes AJBurns MChang CH, et al. ; Medicaid Outcomes Distributed Research Network (MODRN) (2021) Use of medications for treatment of opioid use disorder among US Medicaid enrollees in 11 states, 2014-2018. JAMA 326:154–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Dryden C, Young D, Hepburn M, Mactier H (2009) Maternal methadone use in pregnancy: factors associated with the development of neonatal abstinence syndrome and implications for healthcare resources. BJOG 116:665–671. [DOI] [PubMed] [Google Scholar]
  17. Elkader A, Sproule B (2005) Buprenorphine: clinical pharmacokinetics in the treatment of opioid dependence. Clin Pharmacokinet 44:661–680. [DOI] [PubMed] [Google Scholar]
  18. Harris DS, Mendelson JE, Lin ET, Upton RA, Jones RT (2004) Pharmacokinetics and subjective effects of sublingual buprenorphine, alone or in combination with naloxone: lack of dose proportionality. Clin Pharmacokinet 43:329–340. [DOI] [PubMed] [Google Scholar]
  19. Hassan HE, Myers AL, Coop A, Eddington ND (2009) Differential involvement of P-glycoprotein (ABCB1) in permeability, tissue distribution, and antinociceptive activity of methadone, buprenorphine, and diprenorphine: in vitro and in vivo evaluation. J Pharm Sci 98:4928–4940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Hjelmström P, Banke Nordbeck E, Tiberg F (2020) Optimal dose of buprenorphine in opioid use disorder treatment: a review of pharmacodynamic and efficacy data. Drug Dev Ind Pharm 46:1–7. [DOI] [PubMed] [Google Scholar]
  21. Holland MJ, Carr KD, Simon EJ (1989) Pharmacokinetics of [3H]-buprenorphine in the rat. Res Commun Chem Pathol Pharmacol 64:3–16. [PubMed] [Google Scholar]
  22. Huestis MA, Cone EJ, Pirnay SO, Umbricht A, Preston KL (2013) Intravenous buprenorphine and norbuprenorphine pharmacokinetics in humans. Drug Alcohol Depend 131:258–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Johnson TN, Jamei M, Rowland-Yeo K (2016) How does in vivo biliary elimination of drugs change with age? Evidence from in vitro and clinical data using a systems pharmacology approach. Drug Metab Dispos 44:1090–1098. [DOI] [PubMed] [Google Scholar]
  24. Jones HE, Dengler E, Garrison A, O’Grady KE, Seashore C, Horton E, Andringa K, Jansson LM, Thorp J (2014) Neonatal outcomes and their relationship to maternal buprenorphine dose during pregnancy. Drug Alcohol Depend 134:414–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Jönsson M, Mundin G, Sumner M (2018) Pharmacokinetic and pharmaceutical properties of a novel buprenorphine/naloxone sublingual tablet for opioid substitution therapy versus conventional buprenorphine/naloxone sublingual tablet in healthy volunteers. Eur J Pharm Sci 122:125–133. [DOI] [PubMed] [Google Scholar]
  26. Kalluri HV, Zhang H, Caritis SN, Venkataramanan R (2017) A physiologically based pharmacokinetic modelling approach to predict buprenorphine pharmacokinetics following intravenous and sublingual administration. Br J Clin Pharmacol 83:2458–2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Ko JYD’Angelo DVHaight SCMorrow BCox SSalvesen von Essen BStrahan AEHarrison LTevendale HDWarner L, et al. (2020) Vital signs: prescription opioid pain reliever use during pregnancy - 34 U.S. Jurisdictions, 2019. MMWR Morb Mortal Wkly Rep 69:897–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Kuhlman JJ Jr, Lalani S, Magluilo J Jr, Levine B, Darwin WD (1996) Human pharmacokinetics of intravenous, sublingual, and buccal buprenorphine. J Anal Toxicol 20:369–378. [DOI] [PubMed] [Google Scholar]
  29. Leander JD (1988) Buprenorphine is a potent kappa-opioid receptor antagonist in pigeons and mice. Eur J Pharmacol 151:457–461. [DOI] [PubMed] [Google Scholar]
  30. Lim SCB, Schug S, Krishnarajah J (2019) The pharmacokinetics and local tolerability of a novel sublingual formulation of buprenorphine. Pain Med 20:143–152. [DOI] [PubMed] [Google Scholar]
  31. Liu AJ, Jones MP, Murray H, Cook CM, Nanan R (2010) Perinatal risk factors for the neonatal abstinence syndrome in infants born to women on methadone maintenance therapy. Aust N Z J Obstet Gynaecol 50:253–258. [DOI] [PubMed] [Google Scholar]
  32. Martin WR, Eades CG, Thompson JA, Huppler RE, Gilbert PE (1976) The effects of morphine- and nalorphine-like drugs in the nondependent and morphine-dependent chronic spinal dog. J Pharmacol Exp Ther 197:517–532. [PubMed] [Google Scholar]
  33. McAleer SD, Mills RJ, Polack T, Hussain T, Rolan PE, Gibbs AD, Mullins FG, Hussein Z (2003) Pharmacokinetics of high-dose buprenorphine following single administration of sublingual tablet formulations in opioid naïve healthy male volunteers under a naltrexone block. Drug Alcohol Depend 72:75–83. [DOI] [PubMed] [Google Scholar]
  34. Mendelson J, Upton RA, Everhart ET, Jacob P 3rd, Jones RT (1997) Bioavailability of sublingual buprenorphine. J Clin Pharmacol 37:31–37. [DOI] [PubMed] [Google Scholar]
  35. Mizuno T, McPhail BT, Kamatkar S, Wexelblatt S, Ward L, Christians U, Akinbi HT, Vinks AA (2021) Physiologic indirect response modeling to describe buprenorphine pharmacodynamics in newborns treated for neonatal opioid withdrawal syndrome. Clin Pharmacokinet 60:249–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Moody DE, Fang WB, Morrison J, McCance-Katz E (2011) Gender differences in pharmacokinetics of maintenance dosed buprenorphine. Drug Alcohol Depend 118:479–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Moore JN, Gastonguay MR, Ng CM, Adeniyi-Jones SC, Moody DE, Fang WB, Ehrlich ME, Kraft WK (2018) The pharmacokinetics and pharmacodynamics of buprenorphine in neonatal abstinence syndrome. Clin Pharmacol Ther 103:1029–1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Nath RP, Upton RA, Everhart ET, Cheung P, Shwonek P, Jones RT, Mendelson JE (1999) Buprenorphine pharmacokinetics: relative bioavailability of sublingual tablet and liquid formulations. J Clin Pharmacol 39:619–623. [DOI] [PubMed] [Google Scholar]
  39. Negus SS, Bidlack JM, Mello NK, Furness MS, Rice KC, and Brandt MR (2002) Delta opioid antagonist effects of buprenorphine in rhesus monkeys. Behav Pharmacol 13:557–570. [DOI] [PubMed] [Google Scholar]
  40. Ng CM, Dombrowsky E, Lin H, Erlich ME, Moody DE, Barrett JS, Kraft WK (2015) Population pharmacokinetic model of sublingual buprenorphine in neonatal abstinence syndrome. Pharmacotherapy 35:670–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Patrick SW, Barfield WD, Poindexter BB; Committee on Fetus and Newborn, Committee on Substance Use and Prevention (2020) Neonatal opioid withdrawal syndrome. Pediatrics 146:e2020029074. [DOI] [PubMed] [Google Scholar]
  42. Pergolizzi JAloisi AMDahan AFilitz JLangford RLikar RMercadante SMorlion BRaffa RBSabatowski R, et al. (2010) Current knowledge of buprenorphine and its unique pharmacological profile. Pain Pract 10:428–450. [DOI] [PubMed] [Google Scholar]
  43. Picard N, Cresteil T, Djebli N, Marquet P (2005) In vitro metabolism study of buprenorphine: evidence for new metabolic pathways. Drug Metab Dispos 33:689–695. [DOI] [PubMed] [Google Scholar]
  44. Rodgers T, Rowland M (2006) Physiologically based pharmacokinetic modelling 2: predicting the tissue distribution of acids, very weak bases, neutrals and zwitterions. J Pharm Sci 95:1238–1257. [DOI] [PubMed] [Google Scholar]
  45. Schuh KJ, Johanson CE (1999) Pharmacokinetic comparison of the buprenorphine sublingual liquid and tablet. Drug Alcohol Depend 56:55–60. [DOI] [PubMed] [Google Scholar]
  46. Silva LL, Silvola RM, Haas DM, Quinney SK (2022) Physiologically based pharmacokinetic modelling in pregnancy: Model reproducibility and external validation. Br J Clin Pharmacol 88:1441–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Simon AE, Freund MP, Archer SW, Bremer AA (2021) Toward the use of buprenorphine in infants for neonatal opioid withdrawal syndrome: summary of an NIH workshop. J Perinatol 41:1213–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Strain EC, Moody DE, Stoller KB, Walsh SL, Bigelow GE (2004) Relative bioavailability of different buprenorphine formulations under chronic dosing conditions. Drug Alcohol Depend 74:37–43. [DOI] [PubMed] [Google Scholar]
  49. Takahashi Y, Ishii S, Arizono H, Nishimura S, Tsuruda K, Saito N, Nemoto H, Jin Y, Esumi Y (2001) Pharmacokinetics of buprenorphine hydrochloride (BN•HCl) (1): absorption, distribution, metabolism and excretion after percutaneous (TSN-09: BN•HCl containing tape application) or subcutaneous administration of BN•HCl in rats. Xenobiot Metab Dispos 16:569–583. [Google Scholar]
  50. Upton RN, Semple TJ, Macintyre PE (1997) Pharmacokinetic optimisation of opioid treatment in acute pain therapy. Clin Pharmacokinet 33:225–244. [DOI] [PubMed] [Google Scholar]
  51. van Hoogdalem MW, Johnson TN, McPhail BT, Kamatkar S, Wexelblatt SL, Ward LP, Christians U, Akinbi HT, Vinks AA, and Mizuno T (2022a) Physiologically-based pharmacokinetic modeling to investigate the effect of maturation on buprenorphine pharmacokinetics in newborns with neonatal opioid withdrawal syndrome. Clin Pharmacol Ther 111:496–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. van Hoogdalem MW, McPhail BT, Hahn D, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T (2021) Pharmacotherapy of neonatal opioid withdrawal syndrome: a review of pharmacokinetics and pharmacodynamics. Expert Opin Drug Metab Toxicol 17:87–103. [DOI] [PubMed] [Google Scholar]
  53. van Hoogdalem MW, Tanaka R, Abduljalil K, Johnson TN, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T (2024) Forecasting Fetal Buprenorphine Exposure through Maternal-Fetal Physiologically Based Pharmacokinetic Modeling. Pharmaceutics 16:375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. van Hoogdalem MW, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T (2022b) A review of pregnancy-induced changes in opioid pharmacokinetics, placental transfer, and fetal exposure: Towards fetomaternal physiologically-based pharmacokinetic modeling to improve the treatment of neonatal opioid withdrawal syndrome. Pharmacol Ther 234:108045. [DOI] [PubMed] [Google Scholar]
  55. Wachman EM, Schiff DM, Silverstein M (2018) Neonatal abstinence syndrome: advances in diagnosis and treatment. JAMA 319:1362–1374. [DOI] [PubMed] [Google Scholar]
  56. Wnendt S, Krüger T, Janocha E, Hildebrandt D, Englberger W (1999) Agonistic effect of buprenorphine in a nociceptin/OFQ receptor-triggered reporter gene assay. Mol Pharmacol 56:334–338. [DOI] [PubMed] [Google Scholar]
  57. Yassen A, Olofsen E, Romberg R, Sarton E, Teppema L, Danhof M, Dahan A (2007) Mechanism-based PK/PD modeling of the respiratory depressant effect of buprenorphine and fentanyl in healthy volunteers. Clin Pharmacol Ther 81:50–58. [DOI] [PubMed] [Google Scholar]
  58. Zhang H, Kalluri HV, Bastian JR, Chen H, Alshabi A, Caritis SN, Venkataramanan R (2018) Gestational changes in buprenorphine exposure: A physiologically-based pharmacokinetic analysis. Br J Clin Pharmacol 84:2075–2087. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Drug Metabolism and Disposition are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES