Skip to main content
Log in

Physiologic Indirect Response Modeling to Describe Buprenorphine Pharmacodynamics in Newborns Treated for Neonatal Opioid Withdrawal Syndrome

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objective

Buprenorphine has been shown to be effective in treating infants with neonatal opioid withdrawal syndrome. However, an evidence-based buprenorphine dosing strategy has not been established in the treatment of neonatal opioid withdrawal syndrome because of a lack of exposure–response data. The aim of this study was to develop an integrated pharmacokinetic and pharmacodynamic model to predict buprenorphine treatment outcomes in newborns with neonatal opioid withdrawal syndrome.

Methods

Clinical data were obtained from 19 newborns with a median (range) gestational age of 37 (34–41) weeks enrolled in a pilot pharmacokinetic study of buprenorphine. Sparse blood sampling, comprising three specimens obtained around the second dose of buprenorphine, was performed using heel sticks with dried blood spot technology. Standardized neonatal opioid withdrawal syndrome severity scores (Finnegan scores) were collected every 3–4 h based on symptoms by bedside nursing staff. Mean Finnegan scores were used as a pharmacodynamic marker in the exposure–response modeling. The blood concentration–Finnegan score relationship was described using a physiologic indirect response model with inclusion of natural disease remission.

Results

A total of 52 buprenorphine blood concentrations and 780 mean Finnegan scores were available for the pharmacokinetic/pharmacodynamic modeling and exposure–response analysis. A one-compartment model with first-order absorption adequately described the pharmacokinetic data. The buprenorphine blood concentration at 50% of maximum effect for the inhibition of disease progression was 0.77 ng/mL (95% confidence interval 0.32–1.2). The inclusion of natural disease remission described as a function of postnatal age significantly improved the model fit.

Conclusions

A buprenorphine pharmacokinetic/pharmacodynamic model was successfully developed. The model could facilitate model-informed optimization of the buprenorphine dosing regimen in the treatment of neonatal opioid withdrawal syndrome.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Winkelman TNA, Villapiano N, Kozhimannil KB, Davis MM, Patrick SW. Incidence and costs of neonatal abstinence syndrome among infants with Medicaid: 2004–2014. Pediatrics. 2018;141(4):e20173520.

    Article  Google Scholar 

  2. Patrick SW, Davis MM, Lehmann CU, Cooper WO. Increasing incidence and geographic distribution of neonatal abstinence syndrome: United States 2009 to 2012. J Perinatol. 2015;35(8):650–5.

    Article  CAS  Google Scholar 

  3. McQueen K, Murphy-Oikonen J. Neonatal abstinence syndrome. N Engl J Med. 2016;375(25):2468–79.

    Article  Google Scholar 

  4. Kraft WK, Adeniyi-Jones SC, Chervoneva I, Greenspan JS, Abatemarco D, Kaltenbach K, et al. Buprenorphine for the treatment of the neonatal abstinence syndrome. N Engl J Med. 2017;376(24):2341–8.

    Article  CAS  Google Scholar 

  5. Hall ES, Rice WR, Folger AT, Wexelblatt SL. Comparison of neonatal abstinence syndrome treatment with sublingual buprenorphine versus conventional opioids. Am J Perinatol. 2018;35(4):405–12.

    Article  Google Scholar 

  6. Elkader A, Sproule B. Buprenorphine: clinical pharmacokinetics in the treatment of opioid dependence. Clin Pharmacokinet. 2005;44(7):661–80.

    Article  CAS  Google Scholar 

  7. Hall ES, Meinzen-Derr J, Wexelblatt SL. Cohort analysis of a pharmacokinetic-modeled methadone weaning optimization for neonatal abstinence syndrome. J Pediatr. 2015;167(6):1221–5.e1.

    Article  CAS  Google Scholar 

  8. Wiles JR, Isemann B, Mizuno T, Tabangin ME, Ward LP, Akinbi H, et al. Pharmacokinetics of oral methadone in the treatment of neonatal abstinence syndrome: a pilot study. J Pediatr. 2015;167(6):1214–20.e3.

    Article  CAS  Google Scholar 

  9. Ng CM, Dombrowsky E, Lin H, Erlich ME, Moody DE, Barrett JS, et al. Population pharmacokinetic model of sublingual buprenorphine in neonatal abstinence syndrome. Pharmacotherapy. 2015;35(7):670–80.

    Article  CAS  Google Scholar 

  10. Moore JN, Gastonguay MR, Ng CM, Adeniyi-Jones SC, Moody DE, Fang WB, et al. The pharmacokinetics and pharmacodynamics of buprenorphine in neonatal abstinence syndrome. Clin Pharmacol Ther. 2018;103(6):1029–37.

    Article  CAS  Google Scholar 

  11. Kamatkar S, Mizuno T, Ward LP, Isemann B, Vinks A, Akinbi H. Pharmacokinetics of buprenorphine in the neonatal abstinence syndrome. San Francisco: Pediatric Academic Societies; 2017.

    Google Scholar 

  12. Finnegan LP, Connaughton JF Jr, Kron RE, Emich JP. Neonatal abstinence syndrome: assessment and management. Addict Dis. 1975;2(1):141–58.

    CAS  PubMed  Google Scholar 

  13. Clavijo CF, Hoffman KL, Thomas JJ, Carvalho B, Chu LF, Drover DR, et al. A sensitive assay for the quantification of morphine and its active metabolites in human plasma and dried blood spots using high-performance liquid chromatography-tandem mass spectrometry. Anal Bioanal Chem. 2011;400(3):715–28.

    Article  CAS  Google Scholar 

  14. Lindbom L, Pihlgren P, Jonsson EN. PsN-Toolkit: a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed. 2005;79(3):241–57.

    Article  Google Scholar 

  15. Anderson BJ, Holford NH. Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol. 2008;48:303–32.

    Article  CAS  Google Scholar 

  16. Jusko WJ, Ko HC. Physiologic indirect response models characterize diverse types of pharmacodynamic effects. Clin Pharmacol Ther. 1994;56(4):406–19.

    Article  CAS  Google Scholar 

  17. Nasser AF, Heidbreder C, Gomeni R, Fudala PJ, Zheng B, Greenwald MK. A population pharmacokinetic and pharmacodynamic modelling approach to support the clinical development of RBP-6000, a new, subcutaneously injectable, long-acting, sustained-release formulation of buprenorphine, for the treatment of opioid dependence. Clin Pharmacokinet. 2014;53(9):813–24.

    Article  Google Scholar 

  18. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology: drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157–67.

    Article  CAS  Google Scholar 

  19. Kraft WK. Buprenorphine in neonatal abstinence syndrome. Clin Pharmacol Ther. 2018;103(1):112–9.

    Article  CAS  Google Scholar 

  20. Wachman EM, Schiff DM. Bringing attention to a need for a standardized treatment and weaning protocol for neonatal abstinence syndrome. Transl Pediatr. 2016;5(1):12–5.

    PubMed  PubMed Central  Google Scholar 

  21. Hall ES, Wexelblatt SL, Crowley M, Grow JL, Jasin LR, Klebanoff MA, et al. Implementation of a neonatal abstinence syndrome weaning protocol: a multicenter cohort study. Pediatrics. 2015;136(4):e803–e810810.

    Article  Google Scholar 

  22. Hall ES, Wexelblatt SL, Crowley M, Grow JL, Jasin LR, Klebanoff MA, et al. A multicenter cohort study of treatments and hospital outcomes in neonatal abstinence syndrome. Pediatrics. 2014;134(2):e527–e534534.

    Article  Google Scholar 

  23. Mizuno T, Fukuda T, Emoto C, Mobberley-Schuman PS, Hammill AM, Adams DM, et al. Developmental pharmacokinetics of sirolimus: implications for precision dosing in neonates and infants with complicated vascular anomalies. Pediatr Blood Cancer. 2017. https://doi.org/10.1002/pbc.26470.

    Article  PubMed  Google Scholar 

  24. Mizuno T, Gist KM, Gao Z, Wempe MF, Alten J, Cooper DS, et al. Developmental pharmacokinetics and age-appropriate dosing design of milrinone in neonates and infants with acute kidney injury following cardiac surgery. Clin Pharmacokinet. 2019;58(6):793–803.

    Article  CAS  Google Scholar 

  25. Vinks AA, Emoto C, Fukuda T. Modeling and simulation in pediatric drug therapy: application of pharmacometrics to define the right dose for children. Clin Pharmacol Ther. 2015;98(3):298–308.

    Article  CAS  Google Scholar 

  26. Ohio Perinatal Quality Collaborative. https://www.opqc.net/. Accessed 20 Jan 2020.

  27. Picard N, Cresteil T, Djebli N, Marquet P. In vitro metabolism study of buprenorphine: evidence for new metabolic pathways. Drug Metab Dispos. 2005;33(5):689–95.

    Article  CAS  Google Scholar 

  28. Chang Y, Moody DE, McCance-Katz EF. Novel metabolites of buprenorphine detected in human liver microsomes and human urine. Drug Metab Dispos. 2006;34(3):440–8.

    Article  CAS  Google Scholar 

  29. Rouguieg K, Picard N, Sauvage FL, Gaulier JM, Marquet P. Contribution of the different UDP-glucuronosyltransferase (UGT) isoforms to buprenorphine and norbuprenorphine metabolism and relationship with the main UGT polymorphisms in a bank of human liver microsomes. Drug Metab Dispos. 2010;38(1):40–5.

    Article  CAS  Google Scholar 

  30. Sastre JA, Varela G, Lopez M, Muriel C, Gonzalez-Sarmiento R. Influence of uridine diphosphate-glucuronyltransferase 2B7 (UGT2B7) variants on postoperative buprenorphine analgesia. Pain Pract. 2015;15(1):22–30.

    Article  Google Scholar 

  31. Blanco F, Muriel C, Labrador J, Gonzalez-Porras JR, Gonzalez-Sarmiento R, Lozano FS. Influence of UGT2B7, CYP3A4, and OPRM1 gene polymorphisms on transdermal buprenorphine pain control in patients with critical lower limb ischemia awaiting revascularization. Pain Pract. 2016;16(7):842–9.

    Article  Google Scholar 

  32. Wachman EM, Hayes MJ, Brown MS, Paul J, Harvey-Wilkes K, Terrin N, et al. Association of OPRM1 and COMT single-nucleotide polymorphisms with hospital length of stay and treatment of neonatal abstinence syndrome. JAMA. 2013;309(17):1821–7.

    Article  CAS  Google Scholar 

  33. Grossman MR, Lipshaw MJ, Osborn RR, Berkwitt AK. A novel approach to assessing infants with neonatal abstinence syndrome. Hosp Pediatr. 2018;8(1):1–6.

    Article  Google Scholar 

  34. Blount T, Painter A, Freeman E, Grossman M, Sutton AG. Reduction in length of stay and morphine use for NAS with the "eat, sleep, console" method. Hosp Pediatr. 2019;9(8):615–23.

    Article  Google Scholar 

  35. Grisham LM, Stephen MM, Coykendall MR, Kane MF, Maurer JA, Bader MY. Eat, sleep, console approach: a family-centered model for the treatment of neonatal abstinence syndrome. Adv Neonatal Care. 2019;19(2):138–44.

    Article  Google Scholar 

  36. MacMillan KDL, Rendon CP, Verma K, Riblet N, Washer DB, Volpe HA. Association of rooming-in with outcomes for neonatal abstinence syndrome: a systematic review and meta-analysis. JAMA Pediatr. 2018;172(4):345–51.

    Article  Google Scholar 

Download references

Acknowledgements

We thank Nieko Punt (Medimatics, Maastricht, the Netherlands) for his scientific inputs.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tomoyuki Mizuno.

Ethics declarations

Funding

Brooks T. McPhail was supported by a Eunice Kennedy Shriver National Institute of Child Health and Human Development training Grant (5T32HD069054).

Conflict of interest

Scott Wexelblatt has received a research grant from Chiesi Pharmaceuticals and a consulting fee from Braeburn Pharmaceuticals. He is also on a speaker bureau to discuss treatment options for NOWS. These payments had no effect on this paper or results. Tomoyuki Mizuno, Brooks T. McPhail, Suyog Kamatkar, Laura Ward, Uwe Christians, Henry T. Akinbi, and Alexander A. Vinks have no conflicts of interest that are directly relevant to the content of this article.

Ethics approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Consent to participate

Informed consent was obtained from all individual participants included in the study.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Author contributions

TM, BTM, SK, SW, LW, UC, HTA, and AAV wrote the manuscript and designed the research. TM, BTM, SK, LW, UC, HTA, and AAV performed the research and analyzed the data.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary files (PDF 168 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mizuno, T., McPhail, B.T., Kamatkar, S. et al. Physiologic Indirect Response Modeling to Describe Buprenorphine Pharmacodynamics in Newborns Treated for Neonatal Opioid Withdrawal Syndrome. Clin Pharmacokinet 60, 249–259 (2021). https://doi.org/10.1007/s40262-020-00939-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-020-00939-2

Navigation