1
|
Alshogran OY, Zhao W, Krans EE, Caritis S, Shaik IH, Venkataramanan R. An Approach for Improving the Detection and Quantitation of Buprenorphine and Its Metabolites in Maternal and Neonatal Hair. Ther Drug Monit 2024:00007691-990000000-00302. [PMID: 39671206 DOI: 10.1097/ftd.0000000000001291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/12/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Buprenorphine (BUP) use is prevalent in pregnant women with opioid use disorder (OUD). Drug monitoring during pregnancy is critical for optimizing dosing regimen and achieving the desired clinical outcomes. Hair can be used as a critical biological matrix for monitoring long-term exposure to drugs. The aim of this study was to optimize the methodology used to quantify BUP and its metabolites in hair samples. METHODS Conditions for hair sample processing (ie, hair washing, incubation temperature, and extraction time) were optimized to maximize extraction recovery. The LC-MS/MS strategy employed here used 4 deuterated internal standards for quantifying BUP and its major metabolites [norbuprenorphine (NBUP), buprenorphine glucuronide (BUP-G), and norbuprenorphine-glucuronide (NBUP-G)] in human hair samples. The optimized conditions were used to measure BUP and its metabolites in hair samples of 5 women undergoing OUD treatment and their neonates. RESULTS Unwashed hair samples processed by shaking with acetonitrile for 24 hours at 37 °C showed higher BUP (36%) and NBUP (67%) recovery, compared with those processed by incubation at room temperature. The standard curves showed excellent linearity over 0.05-100 ng/mL for BUP and NBUP and 0.1-200 ng/mL for BUP-G and NBUP-G. The assay was partially validated for reproducibility and accuracy and was successfully used for measuring BUP and metabolites in aforementioned hair samples. BUP was identified in all hair samples, while BUP-G was not. BUP was the primary analyte in maternal hair (median: 38.3 pg/mg; 25-75 percentile: 17-152.4 pg/mg), while NBUP-G was predominant in neonatal hair (median: 28.6 pg/mg; 25%-75% percentile: 1.9-112.8 pg/mg). CONCLUSIONS The methodology used for quantifying BUP and its metabolites in hair samples of maternal female patients and their neonates is simple, accurate, and reproducible. The developed method may be useful for measuring fetal exposure to BUP during gestation.
Collapse
Affiliation(s)
- Osama Y Alshogran
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elizabeth E Krans
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, UPMC Magee-Women's Hospital
- Magee-Women's Research Institute; and
| | - Steve Caritis
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, UPMC Magee-Women's Hospital
- Magee-Women's Research Institute; and
| | - Imam H Shaik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania
- Magee-Women's Research Institute; and
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
2
|
Lynn T, Kelleher ME, Georges HM, McCauley EM, Logan RW, Yonkers KA, Abrahams VM. Buprenorphine Induces Human Fetal Membrane Sterile Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624850. [PMID: 39605446 PMCID: PMC11601656 DOI: 10.1101/2024.11.22.624850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Opioid-use disorder (OUD) during pregnancy has increased in the United States to critical levels and is a leading cause of maternal morbidity and mortality. Untreated OUD is associated with pregnancy complications in particular, preterm birth. Medications for OUD, such as buprenorphine, are recommended with the added benefit that treatment during pregnancy increases treatment post-partum. However, the rate of preterm birth in individuals using illicit opioids or being treated with opioid agonist therapeutics is double that of the general population. Since inflammation in the placenta and the associated fetal membranes (FM) is a common underlying cause of preterm birth, we sought to determine if the opioid, buprenorphine, induces sterile inflammation in human FMs and to examine the mechanisms involved. Using an established in vitro human FM explant system, we report that buprenorphine significantly increased FM secretion of the inflammatory cytokine IL-6; the neutrophilic chemokine IL-8; and the inflammasome-mediated cytokine IL-1β, mirroring the inflammatory profile commonly seen at the maternal-fetal interface in preterm birth. Other factors that were elevated in FMs exposed to buprenorphine included the mediators of membrane weakening, prostaglandin E2 (PGE2), and matrix metalloproteinases, MMP1 and MMP9. Furthermore, this sterile inflammatory and weakening FM response induced by buprenorphine was mediated in part by innate immune Toll-like receptor 4 (TLR4), the NLRP3 inflammasome, the μ-opioid receptor, and downstream NFκB and ERK/JNK/MAPK signaling. This may provide the mechanistic link between opioid use in pregnancy and the elevated risk for preterm birth. Since there are adverse consequences of not treating OUD, our findings may help identify ways to mitigate the impact opioids have on pregnancy outcomes while allowing the continuation of maintenance therapy.
Collapse
|
3
|
Caritis SN, Venkataramanan R. A Pharmacologic Evaluation of Buprenorphine in Pregnancy and the Postpartum Period. J Addict Med 2024:01271255-990000000-00377. [PMID: 39221812 DOI: 10.1097/adm.0000000000001380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
BACKGROUND The dosing regimen in the package insert for sublingual buprenorphine is similar for pregnant and nonpregnant people despite the physiologic changes seen during pregnancy. AIMS To compare plasma buprenorphine pharmacokinetics during and after pregnancy and relate buprenorphine concentration to the pharmacodynamic endpoints of pupil diameter, Clinical Opioid Withdrawal Scale (COWS), and craving scores. STUDY DESIGN Prospective cohort of 22 pregnant people undergoing 33 pharmacologic studies (6-8 hours each) during pregnancy or postpartum. Participants were on a stable daily dose of 2-8 mg sublingual buprenorphine every 6 or 8 hours. The dosing frequency was selected by the participant. On study day, baseline measurements of plasma buprenorphine, pupil diameter, COWS, and craving scores were obtained, then the usual morning dose was taken, and measurements were repeated several times over 1 dosing interval. FINDINGS The dose-normalized area under the plasma buprenorphine concentration time curve was significantly (P = 0.036) lower during pregnancy (155 ± 52 ng × min/mL) than postpartum (218 ± 113 ng × min/mL). Buprenorphine trough concentrations were similar at the start (1.1 ± 0.7 ng/mL) and end of a dosing cycle (1.2 ± 0.8 ng/mL) regardless of dosing frequency. Pupillary diameter, COWS, and craving scores returned to baseline as buprenorphine concentrations approached ~1 ng/mL. CONCLUSIONS Pregnant people require a higher dose of buprenorphine to achieve concentrations comparable to nonpregnant people. There is a temporal relationship between the plasma buprenorphine concentration and the pharmacodynamic markers of pupillary diameter, COWS, and craving scores. An average plasma concentration of ~1 ng/mL was associated with the lowest level of COWS and craving scores.
Collapse
Affiliation(s)
- Steve N Caritis
- From the Departments of Obstetrics and Gynecology and Reproductive Sciences (SNC), Pathology (RV), and Pharmaceutical Sciences (RV), University of Pittsburgh Schools of Medicine and Pharmacy, Pittsburgh, PA
| | | |
Collapse
|
4
|
Jansson LM, McConnell K, Velez ML, Spencer N, Milio L, Leoutsakos J, DiPietro JA. Gestational buprenorphine-naloxone exposure and fetal neurobehavior. Neurotoxicol Teratol 2024; 104:107368. [PMID: 38906389 PMCID: PMC11300143 DOI: 10.1016/j.ntt.2024.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Buprenorphine-naloxone treatment may confer substantial benefits for the treatment of opioid use disorder (OUD) during pregnancy including lower risk for overdose/death, less diversion potential and reduced use of other substances. Treatment may also result in less severe Neonatal Abstinence Syndrome (NAS), but little is known about the effects of this medication on fetal neurodevelopment. METHODS The purpose of the current study is to evaluate neurobehaviors among fetuses exposed to buprenorphine-naloxone at four time points over the second and third trimesters of gestation in pregnant women with OUD on buprenorphine-naloxone therapy. Sixty minutes of continuous fetal monitoring via fetal actocardiograph with a single wide array abdominal transducer took place at times of peak and trough buprenorphine-naloxone levels in 24 pregnant women. Data collection, which included measures of fetal heart rate and motor activity, was conducted between 24 and 36 weeks gestation, with the majority (84.6%) monitored at two or more gestational ages. Medication dose and other substance use was monitored throughout the study and infant NAS severity was assessed. RESULTS Fetal heart rate (FHR), FHR variability, accelerations in FHR, and motor activity were suppressed when buprenorphine-naloxone levels were at pharmacologic peak as compared to trough concentrations at 36 weeks, but not earlier in gestation. Maternal medication dose was unrelated to infant NAS severity. CONCLUSIONS Conclusions: There were evident subclinical fetal neurophysiological responses at times of peak maternal buprenorphine/naloxone levels in later gestation, similar to those previously described for buprenorphine only. Further studies evaluating the effects of these changes in fetal neurobehaviors on the longer-term infant development are needed.
Collapse
Affiliation(s)
- Lauren M Jansson
- Johns Hopkins University School of Medicine, Department of Pediatrics, Baltimore, MD 21224, USA.
| | - Krystle McConnell
- Johns Hopkins University School of Medicine, Department of Pediatrics, Baltimore, MD 21224, USA
| | - Martha L Velez
- Johns Hopkins University School of Medicine, Department of Pediatrics, Baltimore, MD 21224, USA
| | - Nancy Spencer
- Johns Hopkins Bayview Medical Center, Department of Nursing, Baltimore, MD 21224, USA
| | - Lorraine Milio
- Johns Hopkins University School of Medicine, Department of Gynecology and Obstetrics, Baltimore, MD 21224, USA
| | - Jeannie Leoutsakos
- Johns Hopkins University School of Medicine, Department of Psychiatry and Behavioral Sciences, Baltimore, MD 21224, USA
| | - Janet A DiPietro
- Johns Hopkins Bloomberg School of Public Health, Department of Population, Family and Reproductive Health, Baltimore, MD 21224, USA
| |
Collapse
|
5
|
Ferrante JR, Blendy JA. Advances in animal models of prenatal opioid exposure. Trends Neurosci 2024; 47:367-382. [PMID: 38614891 PMCID: PMC11096018 DOI: 10.1016/j.tins.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/20/2024] [Accepted: 03/15/2024] [Indexed: 04/15/2024]
Abstract
Neonatal opioid withdrawal syndrome (NOWS) is a growing public health concern. The complexity of in utero opioid exposure in clinical studies makes it difficult to investigate underlying mechanisms that could ultimately inform early diagnosis and treatments. Clinical studies are unable to dissociate the influence of maternal polypharmacy or the environment from direct effects of in utero opioid exposure, highlighting the need for effective animal models. Early animal models of prenatal opioid exposure primarily used the prototypical opioid, morphine, and opioid exposure that was often limited to a narrow period during gestation. In recent years, the number of preclinical studies has grown rapidly. Newer models utilize both prescription and nonprescription opioids and vary the onset and duration of opioid exposure. In this review, we summarize novel prenatal opioid exposure models developed in recent years and attempt to reconcile results between studies while critically identifying gaps within the current literature.
Collapse
Affiliation(s)
- Julia R Ferrante
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Gersch H, Shah D, Chroust A, Bailey B. Can umbilical cord testing add to maternal urine drug screen for evaluation of infants at risk of neonatal opioid withdrawal syndrome? J Matern Fetal Neonatal Med 2023; 36:2211706. [PMID: 37183045 DOI: 10.1080/14767058.2023.2211706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
OBJECTIVE This study evaluated maternal urine drug screen (UDS) at delivery and umbilical cord drug testing and its association with neonatal opioid withdrawal syndrome (NOWS) diagnosis and severity following opioid exposed pregnancy. METHODS A retrospective chart review of 770 mother-infant dyads at five birthing hospitals in the United States Appalachian region for a five-year period was performed. Variables of interest included dyad demographics, results of maternal UDS at delivery and umbilical cord drug testing, and three neonatal outcomes: NOWS diagnosis, pharmacologic treatment administered for NOWS, and length of hospital stay (LOS) of the newborn. RESULTS Opioid-positivity was between 8.5% and 66.3% based on maternal UDS at delivery or umbilical cord testing. Odds of NOWS diagnosis and increased infant LOS was best associated with opioid detection in maternal UDS alone (OR = 5.62, 95% CI [3.06, 10.33] and OR = 8.33, 95% CI [3.67, 18.89], respectively). However, odds of pharmacologic treatment for NOWS was best associated with opioid detection in both maternal UDS and umbilical cord testing on the same dyad (OR = 3.22, 95% CI [1.14, 9.09]). CONCLUSION Maternal UDS is a better option compared to umbilical cord testing for evaluation of opioid-exposed infants and risk of NOWS diagnosis and increased infant LOS.
Collapse
Affiliation(s)
- Hannah Gersch
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Darshan Shah
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Alyson Chroust
- Department of Psychology, College of Arts and Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Beth Bailey
- Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
- College of Medicine, Central Michigan University, Mt. Pleasant, MI, USA
| |
Collapse
|
7
|
Janganati V, Salazar P, Parks BJ, Gorman GS, Prather PL, Peterson EC, Alund AW, Moran JH, Crooks PA, Brents LK. Deuterated buprenorphine retains pharmacodynamic properties of buprenorphine and resists metabolism to the active metabolite norbuprenorphine in rats. Front Pharmacol 2023; 14:1123261. [PMID: 37229250 PMCID: PMC10204800 DOI: 10.3389/fphar.2023.1123261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/04/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: An active metabolite of buprenorphine (BUP), called norbuprenorphine (NorBUP), is implicated in neonatal opioid withdrawal syndrome when BUP is taken during pregnancy. Therefore, reducing or eliminating metabolism of BUP to NorBUP is a novel strategy that will likely lower total fetal exposure to opioids and thus improve offspring outcomes. Precision deuteration alters pharmacokinetics of drugs without altering pharmacodynamics. Here, we report the synthesis and testing of deuterated buprenorphine (BUP-D2). Methods: We determined opioid receptor affinities of BUP-D2 relative to BUP with radioligand competition receptor binding assays, and the potency and efficacy of BUP-D2 relative to BUP to activate G-proteins via opioid receptors with [35S]GTPγS binding assays in homogenates containing the human mu, delta, or kappa opioid receptors. The antinociceptive effects of BUP-D2 and BUP were compared using the warm-water tail withdrawal assay in rats. Blood concentration versus time profiles of BUP, BUP-D2, and NorBUP were measured in rats following intravenous BUP-D2 or BUP injection. Results: The synthesis provided a 48% yield and the product was ≥99% deuterated. Like BUP, BUP-D2 had sub-nanomolar affinity for opioid receptors. BUP-D2 also activated opioid receptors and induced antinociception with equal potency and efficacy as BUP. The maximum concentration and the area under the curve of NorBUP in the blood of rats that received BUP-D2 were over 19- and 10-fold lower, respectively, than in rats that received BUP. Discussion: These results indicate that BUP-D2 retains key pharmacodynamic properties of BUP and resists metabolism to NorBUP and therefore holds promise as an alternative to BUP.
Collapse
Affiliation(s)
- Venumadhav Janganati
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Paloma Salazar
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Brian J. Parks
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Gregory S. Gorman
- Pharmaceutical Sciences Research Institute, McWhorter School of Pharmacy, Samford University, Birmingham, AL, United States
| | - Paul L. Prather
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Eric C. Peterson
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Jeffery H. Moran
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- PinPoint Testing, LLC., Little Rock, AR, United States
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Lisa K. Brents
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
8
|
Maass C, Schaller S, Dallmann A, Bothe K, Müller D. Considering developmental neurotoxicity in vitro data for human health risk assessment using physiologically-based kinetic modeling: deltamethrin case study. Toxicol Sci 2023; 192:59-70. [PMID: 36637193 PMCID: PMC10025876 DOI: 10.1093/toxsci/kfad007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Developmental neurotoxicity (DNT) is a potential hazard of chemicals. Recently, an in vitro testing battery (DNT IVB) was established to complement existing rodent in vivo approaches. Deltamethrin (DLT), a pyrethroid with a well-characterized neurotoxic mode of action, has been selected as a reference chemical to evaluate the performance of the DNT IVB. The present study provides context for evaluating the relevance of these DNT IVB results for the human health risk assessment of DLT by estimating potential human fetal brain concentrations after maternal exposure to DLT. We developed a physiologically based kinetic (PBK) model for rats which was then translated to humans considering realistic in vivo exposure conditions (acceptable daily intake [ADI] for DLT). To address existing uncertainties, we designed case studies considering the most relevant drivers of DLT uptake and distribution. Calculated human fetal brain concentrations were then compared with the lowest benchmark concentration achieved in the DNT IVB. The developed rat PBK model was validated on in vivo rat toxicokinetic data of DLT over a broad range of doses. The uncertainty based case study evaluation confirmed that repeated exposure to DLT at an ADI level would likely result in human fetal brain concentrations far below the in vitro benchmark. The presented results indicate that DLT concentrations in the human fetal brain are highly unlikely to reach concentrations associated with in vitro findings under realistic exposure conditions. Therefore, the new in vitro DNT results are considered to have no impact on the current risk assessment approach.
Collapse
Affiliation(s)
| | | | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen 51373, Germany
| | - Kathrin Bothe
- Regulatory Toxicology, Research and Development, Bayer AG, CropScience, 40789 Monheim am Rhein, Germany
| | - Dennis Müller
- Regulatory Toxicology, Research and Development, Bayer AG, CropScience, 40789 Monheim am Rhein, Germany
| |
Collapse
|
9
|
Albano GD, La Spina C, Pitingaro W, Milazzo V, Triolo V, Argo A, Malta G, Zerbo S. Intrauterine and Neonatal Exposure to Opioids: Toxicological, Clinical, and Medico-Legal Issues. TOXICS 2023; 11:62. [PMID: 36668788 PMCID: PMC9866828 DOI: 10.3390/toxics11010062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 06/01/2023]
Abstract
Opioids have a rapid transplacental passage (i.e., less than 60 min); furthermore, symptoms characterize the maternal and fetal withdrawal syndrome. Opioid withdrawal significantly impacts the fetus, inducing worse outcomes and a risk of mortality. Moreover, neonatal abstinence syndrome (NAS) follows the delivery, lasts up to 10 weeks, and requires intensive management. Therefore, the prevention and adequate management of NAS are relevant public health issues. This review aims to summarize the most updated evidence in the literature regarding toxicological, clinical, and forensic issues of intrauterine exposure to opioids to provide a multidisciplinary, evidence-based approach for managing such issues. Further research is required to standardize testing and to better understand the distribution of opioid derivatives in each specimen type, as well as the clinically relevant cutoff concentrations in quantitative testing results. A multidisciplinary approach is required, with obstetricians, pediatricians, nurses, forensic doctors and toxicologists, social workers, addiction specialists, and politicians all working together to implement social welfare and social services for the baby when needed. The healthcare system should encourage multidisciplinary activity in this field and direct suspected maternal and neonatal opioid intoxication cases to local referral centers.
Collapse
|
10
|
Liu J, Grewen K, Gao W. Evidence for the Normalization Effects of Medication for Opioid Use Disorder on Functional Connectivity in Neonates with Prenatal Opioid Exposure. J Neurosci 2022; 42:4555-4566. [PMID: 35552232 PMCID: PMC9172285 DOI: 10.1523/jneurosci.2232-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/30/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
Altered functional connectivity has been reported in infants with prenatal exposure to opioids, which significantly interrupts and influences endogenous neurotransmitter/receptor signaling during fetal programming. Better birth outcomes and long-term developmental outcomes are associated with medication for opioid use disorder (MOUD) during pregnancy, but the neural mechanisms underlying these benefits are largely unknown. We aimed to characterize effects of prenatal opioid/other drug exposure (PODE) and the neural basis for the reported beneficial effects of MOUD by examining neonatal brain functional organization. A cohort of 109 human newborns, 42 PODE, 39 with prenatal exposure to drugs excluding opioids (PDE), 28 drug-free controls (males and females) underwent resting-state fMRI at 2 weeks of age. To examine neural effects of MOUD, PODE infants were separated into subgroups based on whether mothers received MOUD (n = 31) or no treatment (n = 11). A novel heatmap analysis was designed to characterize PODE-associated functional connectivity alterations and MOUD-related effects, and permutation testing identified regions of interest with significant effects. PODE neonates showed alterations beyond those associated with PDE, particularly in reward-related frontal-sensory connectivity. MOUD was associated with a significant reduction of PODE-related alterations in key regions of endogenous opioid pathways including limbic and frontal connections. However, significant residual effects in limbic and subcortical circuitry were observed. These findings confirm altered brain functional organization associated with PODE. Importantly, widespread normalization effects associated with MOUD reveal, for the first time, the potential brain basis of the beneficial effects of MOUD on the developing brain and underscore the importance of this treatment intervention for better developmental outcomes.SIGNIFICANCE STATEMENT This is the first study to reveal the potential neural mechanisms underlying the beneficial effects on the neonate brain associated with MOUD during pregnancy. We identified both normalization and residual effects of MOUD on brain functional architecture by directly comparing neonates prenatally exposed to opioids with MOUD and those exposed to opioids but without MOUD. Our findings confirm altered brain functional organization associated with prenatal opioid exposure and demonstrate that although significant residual effects remain in reward circuitry, MOUD confers significant normalization effects on functional connectivity of regions associated with socioemotional development and reward processing. Together, our results highlight the importance of MOUD intervention for better neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Janelle Liu
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, California 90048
- Departments of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Karen Grewen
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Wei Gao
- Cedars-Sinai Biomedical Imaging Research Institute, Los Angeles, California 90048
- Departments of Biomedical Sciences and Imaging, Cedars-Sinai Medical Center, Los Angeles, California 90048
| |
Collapse
|
11
|
Nieto-Estévez V, Donegan JJ, McMahon CL, Elam HB, Chavera TA, Varma P, Berg KA, Lodge DJ, Hsieh J. Buprenorphine Exposure Alters the Development and Migration of Interneurons in the Cortex. Front Mol Neurosci 2022; 15:889922. [PMID: 35600077 PMCID: PMC9115473 DOI: 10.3389/fnmol.2022.889922] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
The misuse of opioids has reached epidemic proportions over the last decade, with over 2.1 million people in the United States suffering from substance use disorders related to prescription opioid pain relievers. This increase in opioid misuse affects all demographics of society, including women of child-bearing age, which has led to a rise in opioid use during pregnancy. Opioid use during pregnancy has been associated with increased risk of obstetric complications and adverse neonatal outcomes, including neonatal abstinence syndrome. Currently, opioid use disorder in pregnant women is treated with long-acting opioid agonists, including buprenorphine. Although buprenorphine reduces illicit opioid use during pregnancy and improves infant outcomes at birth, few long-term studies of the neurodevelopmental consequences have been conducted. The goal of the current experiments was to examine the effects of buprenorphine on the development of the cortex using fetal brain tissue, 3D brain cultures, and rodent models. First, we demonstrated that we can grow cortical and subpallial spheroids, which model the cellular diversity, connectivity, and activity of the developing human brain. Next, we show that cells in the developing human cortex express the nociceptin opioid (NOP) receptor and that buprenorphine can signal through this receptor in cortical spheroids. Using subpallial spheroids to grow inhibitory interneurons, we show that buprenorphine can alter interneuron development and migration into the cortex. Finally, using a rodent model of prenatal buprenorphine exposure, we demonstrate that alterations in interneuron distribution can persist into adulthood. Together, these results suggest that more research is needed into the long-lasting consequences of buprenorphine exposure on the developing human brain.
Collapse
Affiliation(s)
- Vanesa Nieto-Estévez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jennifer J. Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, The University of Texas Health Science Center, San Antonio, TX, United States
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Courtney L. McMahon
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Hannah B. Elam
- Department of Pharmacology and Center for Biomedical Neuroscience, The University of Texas Health Science Center, San Antonio, TX, United States
| | - Teresa A. Chavera
- Department of Pharmacology and Center for Biomedical Neuroscience, The University of Texas Health Science Center, San Antonio, TX, United States
| | - Parul Varma
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Kelly A. Berg
- Department of Pharmacology and Center for Biomedical Neuroscience, The University of Texas Health Science Center, San Antonio, TX, United States
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, The University of Texas Health Science Center, San Antonio, TX, United States
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Jenny Hsieh
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
12
|
van Hoogdalem MW, Johnson TN, McPhail BT, Kamatkar S, Wexelblatt SL, Ward LP, Christians U, Akinbi HT, Vinks AA, Mizuno T. Physiologically-Based Pharmacokinetic Modeling to Investigate the Effect of Maturation on Buprenorphine Pharmacokinetics in Newborns with Neonatal Opioid Withdrawal Syndrome. Clin Pharmacol Ther 2022; 111:496-508. [PMID: 34679189 PMCID: PMC8748288 DOI: 10.1002/cpt.2458] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/15/2021] [Indexed: 02/03/2023]
Abstract
Neonatal opioid withdrawal syndrome (NOWS) is a major public health concern whose incidence has paralleled the opioid epidemic in the United States. Sublingual buprenorphine is an emerging treatment for NOWS, but given concerns about long-term adverse effects of perinatal opioid exposure, precision dosing of buprenorphine is needed. Buprenorphine pharmacokinetics (PK) in newborns, however, is highly variable. To evaluate underlying sources of PK variability, a neonatal physiologically-based pharmacokinetic (PBPK) model of sublingual buprenorphine was developed using Simcyp (version 19.1). The PBPK model included metabolism by cytochrome P450 (CYP) 3A4, CYP2C8, UDP-glucuronosyltransferase (UGT) 1A1, UGT1A3, UGT2B7, and UGT2B17, with additional biliary excretion. Maturation of metabolizing enzymes was incorporated, and default CYP2C8 and UGT2B7 ontogeny profiles were updated according to recent literature. A biliary clearance developmental profile was outlined using clinical data from neonates receiving sublingual buprenorphine as NOWS treatment. Extensive PBPK model validation in adults demonstrated good predictability, with geometric mean (95% confidence interval (CI)) predicted/observed ratios (P/O ratios) of area under the curve from zero to infinity (AUC0-∞ ), peak concentration (Cmax ), and time to reach peak concentration (Tmax ) equaling 1.00 (0.74-1.33), 1.04 (0.84-1.29), and 0.95 (0.72-1.26), respectively. In neonates, the geometric mean (95% CI) P/O ratio of whole blood concentrations was 0.75 (95% CI 0.64-0.87). PBPK modeling and simulation demonstrated that variability in biliary clearance, sublingual absorption, and CYP3A4 abundance are likely important drivers of buprenorphine PK variability in neonates. The PBPK model could be used to guide development of improved buprenorphine starting dose regimens for the treatment of NOWS.
Collapse
Affiliation(s)
- Matthijs W. van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | | | - Brooks T. McPhail
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- School of Medicine Greenville, University of South Carolina, Greenville, SC, USA
| | - Suyog Kamatkar
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Community Hospital East, Indianapolis, IN, USA
| | - Scott L. Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Laura P. Ward
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Uwe Christians
- iC42 Clinical Research and Development, University of Colorado, Aurora, CO, USA
| | - Henry T. Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alexander A. Vinks
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
13
|
van Hoogdalem MW, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T. 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 2021; 234:108045. [PMID: 34813863 DOI: 10.1016/j.pharmthera.2021.108045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling has emerged as a useful tool to study pharmacokinetics (PK) in special populations, such as pregnant women, fetuses, and newborns, where practical hurdles severely limit the study of drug behavior. PK in pregnant women is variable and everchanging, differing greatly from that in their nonpregnant female and male counterparts typically enrolled in clinical trials. PBPK models can accommodate pregnancy-induced physiological and metabolic changes, thereby providing mechanistic insights into maternal drug disposition and fetal exposure. Fueled by the soaring opioid epidemic in the United States, opioid use during pregnancy continues to rise, leading to an increased incidence of neonatal opioid withdrawal syndrome (NOWS). The severity of NOWS is influenced by a complex interplay of extrinsic and intrinsic factors, and varies substantially between newborns, but the extent of prenatal opioid exposure is likely the primary driver. Fetomaternal PBPK modeling is an attractive approach to predict in utero opioid exposure. To facilitate the development of fetomaternal PBPK models of opioids, this review provides a detailed overview of pregnancy-induced changes affecting the PK of commonly used opioids during gestation. Moreover, the placental transfer of these opioids is described, along with their disposition in the fetus. Lastly, the implementation of these factors into PBPK models is discussed. Fetomaternal PBPK modeling of opioids is expected to provide improved insights in fetal opioid exposure, which allows for prediction of postnatal NOWS severity, thereby opening the way for precision postnatal treatment of these vulnerable infants.
Collapse
Affiliation(s)
- Matthijs W van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Scott L Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Henry T Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
14
|
Histological Changes Observed in Placentas Exposed to Medication-Assisted Treatment. J Pregnancy 2021; 2021:2175026. [PMID: 34659831 PMCID: PMC8516570 DOI: 10.1155/2021/2175026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/06/2021] [Accepted: 09/11/2021] [Indexed: 11/22/2022] Open
Abstract
Introduction To compare the effects of medication-assisted treatment on the placenta in pregnant women with opioid use disorder and uncomplicated pregnancies. Methods This is a case-controlled study of pregnant women utilizing medication-assisted treatment, buprenorphine or methadone, which were matched to healthy uncomplicated controls by gestational age. Placental evaluations and neonatal outcomes were evaluated. Data analysis performed standard statistics and relative risk analysis with a p < 0.05 considered significant. Results There were 143 women who met the inclusion criteria: 103 utilizing MAT, 41 buprenorphine and 62 methadone, and 40 uncomplicated matched healthy controls. The incidence of delayed villous maturation was 36% in the medication-assisted group compared with 10% in controls (RR 3.6: 95% CI 1.37-9.43; p < 0.01). The placental weight was greater (541 ± 117 g versus 491 ± 117 g; p = 0.02), and the fetoplacental weight ratio was lower (5.70 ± 1.1 versus 7.13 ± 1.4; p < 0.01) in the medication-exposed pregnancies compared with controls. The mean birth weight of the MAT newborns was significantly lower than that of the healthy controls (3018 ± 536 g versus 3380 ± 492 g; p < 0.01). When evaluating the subgroups of the MAT newborns, the birth weight of the methadone-exposed newborns (2886 ± 514 g) was significantly lower than that of the buprenorphine-exposed newborns (3218 ± 512 g; p < 0.01). Conclusion Medication-exposed pregnancies have a greater incidence of delayed villous maturation, a larger placental size, and a decreased fetoplacental weight ratio compared to the healthy controls. Larger long-term follow-up studies to evaluate outcomes with the presence of delayed villous maturation are needed.
Collapse
|
15
|
Merhar SL, Kline JE, Braimah A, Kline-Fath BM, Tkach JA, Altaye M, He L, Parikh NA. Prenatal opioid exposure is associated with smaller brain volumes in multiple regions. Pediatr Res 2021; 90:397-402. [PMID: 33177677 PMCID: PMC8110593 DOI: 10.1038/s41390-020-01265-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/02/2022]
Abstract
BACKGROUND The impact of prenatal opioid exposure on brain development remains poorly understood. METHODS We conducted a prospective study of term-born infants with and without prenatal opioid exposure. Structural brain MRI was performed between 40 and 48 weeks postmenstrual age. T2-weighted images were processed using the Developing Human Connectome Project structural pipeline. We compared 63 relative regional brain volumes between groups. RESULTS Twenty-nine infants with prenatal opioid exposure and 42 unexposed controls were included. The groups had similar demographics, except exposed infants had lower birth weights, more maternal smoking and maternal Hepatitis C, fewer mothers with a college degree, and were more likely non-Hispanic White. After controlling for sex, postmenstrual age at scan, birth weight, and maternal education, exposed infants had significantly smaller relative volumes of the deep gray matter, bilateral thalamic ventrolateral nuclei, bilateral insular white matter, bilateral subthalamic nuclei, brainstem, and cerebrospinal fluid. Exposed infants had larger relative volumes of the right cingulate gyrus white matter and left occipital lobe white matter. CONCLUSIONS Infants with prenatal opioid exposure had smaller brain volumes in multiple regions compared to controls, with two regions larger in the opioid-exposed group. Further research should focus on the relative contributions of maternal opioids and other exposures. IMPACT Prenatal opioid exposure is associated with developmental and behavioral consequences, but the direct effects of opioids on the developing human brain are poorly understood. Prior small studies using MRI have shown smaller regional brain volumes in opioid-exposed infants and children. After controlling for covariates, infants with prenatal opioid exposure scanned at 40-48 weeks postmenstrual age had smaller brain volumes in multiple regions compared to controls, with two regions larger in the opioid-exposed group. This adds to the literature showing potential impact of prenatal opioid exposure on the developing brain.
Collapse
Affiliation(s)
- Stephanie L Merhar
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA.
| | - Julia E Kline
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Adebayo Braimah
- Imaging Research Center, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Beth M Kline-Fath
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jean A Tkach
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Mekibib Altaye
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Lili He
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Nehal A Parikh
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
16
|
Concheiro M, Gutierrez FM, Ocampo A, Lendoiro E, González-Colmenero E, Concheiro-Guisán A, Peñas-Silva P, Macías-Cortiña M, Cruz-Landeira A, López-Rivadulla M, de-Castro-Ríos A. Assessment of biological matrices for the detection of in utero cannabis exposure. Drug Test Anal 2021; 13:1371-1382. [PMID: 33750017 DOI: 10.1002/dta.3034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 01/01/2023]
Abstract
Cannabis consumption has been increasing worldwide among pregnant women. Due to the negative effects of prenatal cannabis exposure, it is necessary to develop an objective, sensitive, and specific method to determine cannabinoids use during pregnancy. In this study, we compared four different biological samples, maternal hair, meconium, umbilical cord, and placenta, for the detection of in utero cannabis exposure. The biological samples were collected from 627 mother-newborn dyads. All hair and meconium samples were analyzed, and umbilical cord and placenta if hair and/or meconium were positive for cannabinoids. Meconium and hair showed to complement each other, with an agreement between hair and meconium results of 96.7% but only 34.3% if just positive results were considered. Umbilical cord and placenta results showed a better agreement with meconium (91.3% and 92.6%, respectively) than with hair (39.1% and 34.6%, respectively). The predominant metabolites in meconium were 11-nor-carboxy-THC (THCCOOH) and 8,11-dihydroxy-THC (diOHTHC), and in umbilical cord and placenta was THCCOOH-glucuronide. Cannabidiol (CBD) and cannabinol (CBN) were detected in meconium but not in any umbilical cord or placenta. For the first time, prenatal marijuana exposure was analyzed and compared in paired hair, meconium, umbilical cord, and placental samples. Hair and meconium positivity rate was similar, but a more sensitive and specific analytical method for the hair may resolve discrepancies between the matrices. Umbilical cord and placenta may be considered suitable alternative matrices to meconium through the determination of THCCOOH-glucuronide as a biomarker of cannabis exposure.
Collapse
Affiliation(s)
- Marta Concheiro
- John Jay College of Criminal Justice, City University of New York, New York, NY, USA
| | | | - Alejandro Ocampo
- John Jay College of Criminal Justice, City University of New York, New York, NY, USA
| | - Elena Lendoiro
- Sección de Toxicología, Instituto de Ciencias Forenses, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | - Ana Concheiro-Guisán
- Sección de Neonatología, Complejo Hospitalario Universitario de Vigo, Vigo, Spain
| | - Patricia Peñas-Silva
- Sección de Ginecología y Obstetricia, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Macías-Cortiña
- Sección de Ginecología y Obstetricia, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Angelines Cruz-Landeira
- Sección de Toxicología, Instituto de Ciencias Forenses, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel López-Rivadulla
- Sección de Toxicología, Instituto de Ciencias Forenses, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana de-Castro-Ríos
- Sección de Toxicología, Instituto de Ciencias Forenses, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
17
|
Abstract
PURPOSE Buprenorphine and methadone are international gold standards for managing opioid use disorders. Although they are efficacious in treating opioid dependence, buprenorphine and methadone present risks, especially during pregnancy, causing neonatal abstinence syndrome and adverse obstetrical outcomes. Buprenorphine and methadone are also abused during pregnancy, and identifying their use is important to limit unprescribed prenatal exposure. Previous studies have suggested that concentrations of buprenorphine, but not methadone markers in unconventional matrices may predict child outcomes, although currently only limited data exist. We reviewed the literature on concentrations of buprenorphine, methadone, and their metabolites in unconventional matrices to improve data interpretation. METHODS A literature search was conducted using scientific databases (PubMed, Scopus, Web of Science, and reports from international institutions) to review published articles on buprenorphine and methadone monitoring during pregnancy. RESULTS Buprenorphine and methadone and their metabolites were quantified in the meconium, umbilical cord, placenta, and maternal and neonatal hair. Methadone concentrations in the meconium and hair were typically higher than those in other matrices, although the concentrations in the placenta and umbilical cord were more suitable for predicting neonatal outcomes. Buprenorphine concentrations were lower and required sensitive instrumentation, as measuring buprenorphine glucuronidated metabolites is critical to predict neonatal outcomes. CONCLUSIONS Unconventional matrices are good alternatives to conventional ones for monitoring drug exposure during pregnancy. However, data are currently scarce on buprenorphine and methadone during pregnancy to accurately interpret their concentrations. Clinical studies should be conducted with larger cohorts, considering confounding factors such as illicit drug co-exposure.
Collapse
|
18
|
Coulson CC, Lorencz E, Rittenhouse K, Ramage M, Lorenz K, Galvin SL. Association of Maternal Buprenorphine or Methadone Dose with Fetal Growth Indices and Neonatal Abstinence Syndrome. Am J Perinatol 2021; 38:28-36. [PMID: 31421639 DOI: 10.1055/s-0039-1694729] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE Our objective was to compare fetal growth and incidence of neonatal abstinence syndrome requiring treatment across pregnant women with opioid use disorders on two types and two dose categories of medication-assisted treatment. STUDY DESIGN A retrospective cohort study was conducted in a comprehensive, perinatal program in western North Carolina comparing growth percentiles on third-trimester ultrasound and at birth, and diagnosis of neonatal abstinence syndrome requiring treatment. Singletons were exposed in utero to low- to moderate-dose buprenorphine (≤16 mg/day; n = 70), high-dose buprenorphine (≥17 mg/day; n = 36), low- to moderate-dose methadone (≤89 mg/day; n = 41), or high-dose methadone (≥90 mg/day; n = 74). Multivariate analysis of variance with posthoc Bonferroni comparisons (p ≤ 0.01) and multinomial logistic regressions (adjusted odds ratio, 99% confidence interval) were conducted. RESULTS Differences in neonatal outcomes reached statistical significance for larger head circumference for buprenorphine doses (p = 0.01) and for longer length (p < 0.01) and lower odds of neonatal abstinence syndrome requiring treatment (p < 0.01) with low- to moderate-dose buprenorphine versus high-dose methadone. CONCLUSION Among pregnant women using medication-assisted treatment for opioid use disorders, low- to moderate-dose buprenorphine (≤16 mg/day) was associated with the most favorable neonatal outcomes. However, more rigorous control of confounders with a larger sample is necessary to determine if low- to moderate-dose buprenorphine is the better treatment choice.
Collapse
Affiliation(s)
- Carol C Coulson
- Department of Obstetrics and Gynecology, Mountain Area Health Education Center, Asheville, North Carolina.,Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Erin Lorencz
- Department of Obstetrics and Gynecology, Mountain Area Health Education Center, Asheville, North Carolina
| | - Katelyn Rittenhouse
- University of North Carolina School of Medicine-Asheville, Asheville, North Carolina
| | - Melinda Ramage
- Department of Obstetrics and Gynecology, Mountain Area Health Education Center, Asheville, North Carolina
| | - Kathleen Lorenz
- Department of Obstetrics and Gynecology, Mountain Area Health Education Center, Asheville, North Carolina
| | - Shelley L Galvin
- Department of Obstetrics and Gynecology, Mountain Area Health Education Center, Asheville, North Carolina.,Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
19
|
Martin CE, Shadowen C, Thakkar B, Oakes T, Gal TS, Moeller FG. Buprenorphine dosing for the treatment of opioid use disorder through pregnancy and postpartum. CURRENT TREATMENT OPTIONS IN PSYCHIATRY 2020; 7:375-399. [PMID: 33585165 PMCID: PMC7880143 DOI: 10.1007/s40501-020-00221-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Opioid-related deaths are a leading cause of mortality during pregnancy through 12 months postpartum. Buprenorphine use during pregnancy is increasing, yet expert opinion on its dosing through the perinatal period is limited. We provide a review of the current clinical literature on buprenorphine dosing during pregnancy through 12 months postpartum. and present data from a retrospective chart review of patients at our institution describing trends in buprenorphine dosing during pregnancy and postpartum. Utilizing this information, we synthesize findings to provide clinical recommendations for providers. RECENT FINDINGS Existing literature during pregnancy reflects how many women increase and split total daily buprenorphine doses as gestational age advances. SUMMARY We present data from a retrospective chart review of patients at our institution describing trends in buprenorphine dosing during pregnancy and postpartum. Utilizing this information, we synthesize findings to provide clinical recommendations for providers. Changes in the total daily dose of buprenorphine used across pregnancy and through 12 months postpartum at the individual level do not follow consistent patterns, highlighting substantial individual variability. Altogether, buprenorphine dosing should be individualized through pregnancy and postpartum with frequent evaluations by providers and solicited input from patients.
Collapse
Affiliation(s)
- Caitlin E. Martin
- Virginia Commonwealth University, (Department of Obstetrics & Gynecology, Institute for Drug and Alcohol Studies), Richmond, (Virginia), USA
| | - Caroline Shadowen
- Virginia Commonwealth University, (School of Medicine), Richmond, (Virginia), USA
| | - Bhushan Thakkar
- Virginia Commonwealth University, (Department of Obstetrics & Gynecology), Richmond, (Virginia), USA
| | - Travis Oakes
- Virginia Commonwealth University, (Clinical Research Informatics Group, C. Kenneth and Dianne Wright Center for Clinical and Translational Research), Richmond, (Virginia), USA
| | - Tamas S. Gal
- Virginia Commonwealth University, (Department of Biostatistics, School of Medicine), Richmond, (Virginia), USA
| | - F. Gerard Moeller
- Virginia Commonwealth University, (Department of Psychiatry, Institute for Drug and Alcohol Studies), Richmond, (Virginia), USA
| |
Collapse
|
20
|
Blanco-Castañeda R, Galaviz-Hernández C, Souto PCS, Lima VV, Giachini FR, Escudero C, Damiano AE, Barragán-Zúñiga LJ, Martínez-Aguilar G, Sosa-Macías M. The role of xenobiotic-metabolizing enzymes in the placenta: a growing research field. Expert Rev Clin Pharmacol 2020; 13:247-263. [PMID: 32129110 DOI: 10.1080/17512433.2020.1733412] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The placenta is a temporary and unique organ that allows for the physical connection between a mother and fetus; this organ regulates the transport of gases and nutrients mediating the elimination of waste products contained in the fetal circulation. The placenta performs metabolic and excretion functions, on the basis of multiple enzymatic systems responsible for the oxidation, reduction, hydrolysis, and conjugation of xenobiotics. These mechanisms give the placenta a protective role that limits the fetal exposure to harmful compounds. During pregnancy, some diseases require uninterrupted treatment even if it is detrimental to the fetus. Drugs and other xenobiotics alter gene expression in the placenta with repercussions for the fetus and mother's well-being.Areas covered: This review provides a brief description of the human placental structure and function, the main drug and xenobiotic transporters and metabolizing enzymes, placenta-metabolized substrates, and alterations in gene expression that the exposure to xenobiotics may cause.Expert opinion: Research should be focused on the identification and validation of biological markers for the assessment of the harmful effects of some drugs in pregnancy, including the evaluation of polymorphisms and methylation patterns in chorionic villous samples and/or amniotic fluid.
Collapse
Affiliation(s)
| | | | - Paula C S Souto
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Victor Vitorino Lima
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Fernanda R Giachini
- Laboratory of Vascular Biology, Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra Do Garcas, Brazil
| | - Carlos Escudero
- Vascular Physiology Laboratory Group of Investigation in Tumor Angiogenesis (GIANT) Group of Research and Innovation in Vascular Health (GRIVAS Health) Basic Sciences Department Faculty of Sciences, Universidad Del Bio-Bio, Chillan, Chile
| | - Alicia E Damiano
- Laboratorio De Biología De La Reproducción, IFIBIO Houssay-UBA-CONICET, Buenos Aires, Argentina.,Departamento De Ciencias Biológicas, Facultad De Farmacia Y Bioquimica, Buenos Aires, UBA, Argentina
| | | | - Gerardo Martínez-Aguilar
- Unidad De Investigación Biomédica - Instituto Mexicano del Seguro Social (IMSS) Durango, Durango, México
| | - Martha Sosa-Macías
- Academia De Genómica, Instituto Politécnico Nacional-CIIDIR Durango, Durango, Mexico
| |
Collapse
|
21
|
Chhibber T, Bagchi S, Lahooti B, Verma A, Al-Ahmad A, Paul MK, Pendyala G, Jayant RD. CNS organoids: an innovative tool for neurological disease modeling and drug neurotoxicity screening. Drug Discov Today 2020; 25:456-465. [PMID: 31783130 PMCID: PMC7039749 DOI: 10.1016/j.drudis.2019.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/28/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
The paradigm of central nervous system (CNS) drug discovery has mostly relied on traditional approaches of rodent models or cell-based in vitro models. Owing to the issues of species differences between humans and rodents, it is difficult to correlate the robustness of data for neurodevelopmental studies. With advances in the stem-cell field, 3D CNS organoids have been developed and explored owing to their resemblance to the human brain architecture and functions. Further, CNS organoids provide a unique opportunity to mimic the human brain physiology and serve as a modeling tool to study the normal versus pathological brain or the elucidation of mechanisms of neurological disorders. Here, we discuss the recent application of a CNS organoid explored for neurodevelopment disease or a screening tool for CNS drug development.
Collapse
Affiliation(s)
- Tanya Chhibber
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA
| | - Sounak Bagchi
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA
| | - Behnaz Lahooti
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA
| | - Angela Verma
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA
| | - Manash K Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, College of Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA
| | - Rahul Dev Jayant
- Department of Pharmaceutical Sciences, JH School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX 79106, USA.
| |
Collapse
|
22
|
Caritis SN, Panigrahy A. Opioids affect the fetal brain: reframing the detoxification debate. Am J Obstet Gynecol 2019; 221:602-608. [PMID: 31323217 DOI: 10.1016/j.ajog.2019.07.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/26/2019] [Accepted: 07/12/2019] [Indexed: 10/26/2022]
Abstract
Medication-assisted treatment is recommended for individuals with an opioid use disorder, including pregnant women. Medication-assisted treatment during pregnancy provides benefits to the mother and fetus, including better pregnancy outcomes, reduced illicit drug use, and improved prenatal care. An alternative approach, medically supervised withdrawal (detoxification), has, in recent reports, demonstrated a low risk of fetal death and low rates of relapse and neonatal abstinence syndrome. The rates of relapse and neonatal abstinence syndrome are questioned by many who view medically supervised withdrawal as unacceptable based on the concern for the potential adverse consequences of relapse to mother and baby. The impact of opioids on the fetal brain have not been integrated into this debate. Studies in animals and human brain tissues demonstrate opioid receptors in neurons, astroglia, and oligodendrocytes. Age-specific normative data from infants, children, and adults have facilitated investigation of the impact of opioids on the human brain in vivo. Collectively, these studies in animals, human neural tissue, adult brains, and the brains of children and newborns demonstrate that opioids adversely affect the human brain, primarily the developing oligodendrocyte and the processes of myelinization (white matter microstructure), connectivity between parts of the brain, and the size of multiple brain regions, including the basal ganglia, thalamus, and cerebellar white matter. These in vivo studies across the human lifespan suggest vulnerability of specific fronto-temporal-limbic and frontal-subcortical (basal ganglia and cerebellum) pathways that are also likely vulnerable in the human fetal brain. The long-term impact of these reproducible changes in the fetal brain in vivo is unclear, but the possibility of lasting injury has been suggested. In light of the recent data on medically supervised withdrawal and the emerging evidence suggesting adverse effects of opioids on the developing fetal brain, a new paradigm of care is needed that includes the preferred option of medication-assisted treatment but also the option of medically supervised opioid withdrawal for a select group of women. Both these treatment options should offer mental health and social services support throughout pregnancy. More research on both opioid exposure on the developing human brain and the impact of medically supervised withdrawal is required to identify appropriate candidates, optimal dose reduction regimens, and gestational age timing for initiating medically supervised withdrawal.
Collapse
|
23
|
Kongstorp M, Bogen IL, Stiris T, Andersen JM. High Accumulation of Methadone Compared with Buprenorphine in Fetal Rat Brain after Maternal Exposure. J Pharmacol Exp Ther 2019; 371:130-137. [PMID: 31358559 DOI: 10.1124/jpet.119.259531] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Experimental animal studies are valuable in revealing a causal relationship between prenatal exposure to opioid maintenance treatment (OMT) and subsequent effects; however, previous animal studies of OMT during pregnancy have been criticized for their lack of clinical relevance because of their use of high drug doses and the absence of pharmacokinetic data. Hence, the aim of this study was to determine blood and brain concentrations in rat dams, fetuses, and offspring after continuous maternal exposure to methadone or buprenorphine during gestation and to examine the offspring for neonatal outcomes and withdrawal symptoms. Female rats were implanted with a 28-day osmotic minipump delivering methadone (10 mg/kg per day), buprenorphine (1 mg/kg per day) or vehicle 5 days before mating. Continuous exposure to methadone or buprenorphine induced stable blood concentrations in the dams of 0.25 ± 0.02 µM and 5.65 ± 0.16 nM, respectively. The fetal brain concentration of methadone (1.89 ± 0.35 nmol/g) was twice as high as that in the maternal brain, whereas the fetal brain concentration of buprenorphine (20.02 ± 4.97 pmol/g) was one-third the maternal brain concentration. The opioids remained in the offspring brain several days after the exposure ceased. Offspring prenatally exposed to methadone, but not buprenorphine, displayed reduced body weight and length and increased corticosterone levels. No significant changes in ultrasonic vocalizations were revealed. Our data in rat fetuses and neonates indicate that OMT with buprenorphine may be a better choice than methadone during pregnancy. SIGNIFICANCE STATEMENT: Concern has been raised about the use of opioid maintenance treatment during pregnancy because of the important role of the endogenous opioid system in brain development. Here, we show that the methadone concentration in the fetal rat brain was twice as high as that in the maternal brain, whereas the buprenorphine concentration was one-third the maternal concentration. Furthermore, buprenorphine allowed more favorable birth outcomes, suggesting that buprenorphine may be a better choice during pregnancy.
Collapse
Affiliation(s)
- Mette Kongstorp
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Inger Lise Bogen
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Tom Stiris
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| | - Jannike Mørch Andersen
- Section for Drug Abuse Research, Department of Forensic Sciences (M.K., I.L.B., J.M.A.) and Department of Neonatal Intensive Care (T.S.), Oslo University Hospital, and Institute of Clinical Medicine, Faculty of Medicine (M.K., T.S.), Department of Pharmacy, Faculty of Mathematics and Natural Sciences (J.M.A.), and Institute of Basic Medical Sciences, Faculty of Medicine (I.L.B.), University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Bolin EH, Escalona-Vargas D, Daily JA, Siegel ER, Lowery CL, Coker J, Stowe ZN, Eswaran H. Magnetocardiographic identification of prolonged fetal corrected QT interval in women receiving treatment for opioid use disorder. J Obstet Gynaecol Res 2019; 45:1989-1996. [PMID: 31297963 DOI: 10.1111/jog.14055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/23/2019] [Indexed: 01/04/2023]
Abstract
AIM Pregnant women undergoing treatment for opioid use disorder (OUD) may be exposed to multiple QT prolonging agents. We used magnetocardiography to measure fetal QT intervals in mothers with OUD on buprenorphine therapy. METHODS Fetal and maternal magnetocardiography was performed in pregnant women receiving buprenorphine-assisted treatment (Disorder group); these were matched by gestational age to pregnant women who were opiate naïve (Reference group). Corrected QT intervals were determined using Bazett's formula and compared between groups. RESULTS A total of eight women in the Disorder group matched to eight in the Reference group. Seven of the mothers (88%) in the Disorder group were smokers; there were no smokers in the Reference group. The average fetal corrected QT was significantly longer (P = 0.022) in the Disorder group than that in the Reference group (505 milliseconds [ms] ± 68.6 [standard deviation] vs 383 ms ± 70.3 [standard deviation]). CONCLUSION Novel data from this small sample demonstrate prolongation of fetal corrected QT in women with OUD participating in buprenorphine assisted therapy. Additional investigation from a larger sample is needed to clarify if fetal buprenorphine and/or tobacco exposure is associated with changes in fetal QT which would warrant further prenatal and postnatal testing.
Collapse
Affiliation(s)
- Elijah H Bolin
- Department of Pediatrics, Division of Cardiology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, Arkansas, USA
| | - Diana Escalona-Vargas
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Joshua A Daily
- Department of Pediatrics, Division of Cardiology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, Arkansas, USA
| | - Eric R Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Curtis L Lowery
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jessica Coker
- Department of Psychiatry, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Zachary N Stowe
- Department of Psychiatry, University of Wisconsin at Madison, Madison, Wisconsin, USA
| | - Hari Eswaran
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
25
|
|
26
|
Abstract
Neonatal abstinence syndrome refers to the signs and symptoms attributed to the cessation of prenatal exposure (via placental transfer) to various substances. This Primer focuses on neonatal abstinence syndrome caused by opioid use during pregnancy - neonatal opioid withdrawal syndrome (NOWS). As the global prevalence of opioid use has alarmingly increased, so has the incidence of NOWS. NOWS can manifest with varying severity or not at all, for unknown reasons, but is likely to be associated with multiple factors, both maternal (for example, smoking and additional substance exposures) and neonatal (gestational age, sex and genetics). Care for the infant with NOWS begins with addressing the issues experienced by pregnant women with opioid use disorder. Co-occurring mental illness, economic hardship, intimate partner violence, infectious diseases and limited access to care are common in these women and can result in poor maternal and neonatal outcomes. Although there is no consensus regarding optimal NOWS management, non-pharmacological interventions (such as breastfeeding and rooming-in of the mother and the baby) have become a priority, as they can ameliorate symptoms without the need for further opioid exposure. Untreated NOWS can be associated with morbidity in early infancy, and the long-term consequences of fetal opioid exposure are only beginning to be understood.
Collapse
Affiliation(s)
- Mara G Coyle
- Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, RI, USA.
| | - Susan B Brogly
- Department of Surgery, Queen's University, Kingston, Ontario, Canada
| | - Mahmoud S Ahmed
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Stephen W Patrick
- Vanderbilt Center for Child Health Policy, Department of Pediatrics and Health Policy, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hendrée E Jones
- Department of Obstetrics and Gynecology, University of North Carolina, Carrboro, NC, USA
| |
Collapse
|
27
|
Does Maternal Buprenorphine Dose Affect Severity or Incidence of Neonatal Abstinence Syndrome? J Addict Med 2018; 12:435-441. [DOI: 10.1097/adm.0000000000000427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Price HR, Collier AC, Wright TE. Screening Pregnant Women and Their Neonates for Illicit Drug Use: Consideration of the Integrated Technical, Medical, Ethical, Legal, and Social Issues. Front Pharmacol 2018; 9:961. [PMID: 30210343 PMCID: PMC6120972 DOI: 10.3389/fphar.2018.00961] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/03/2018] [Indexed: 01/01/2023] Open
Abstract
North America is currently suffering from one of the worst epidemics of illicit drug use in recent history: the opioid crisis. Pregnant women are not immune to the ravages of substance misuse which affects themselves, their pregnancies, and the wider community. The prevalence of drug misuse in pregnancy is not well quantified due to the lack of good validated tests, cooperation between clinicians and scientists developing tests, and consensus as to who should be tested and how results should be used. A wide range of tissues can be tested for drug use, including maternal blood, urine, and hair; neonatal meconium, urine, and hair; and placenta and umbilical cord tissues. Testing methods range from simple spectrophotometry and clinical chemistry to sophisticated analytical HPLC or mass spectrometry techniques. The drive for ever greater accuracy and sensitivity must be balanced with the necessities of medical practice requiring minimally invasive sampling, rapid turnaround, and techniques that can be realistically utilized in a clinical laboratory. Better screening tests have great potential to improve neonatal and maternal medical outcomes by enhancing the speed and accuracy of diagnosis. They also have great promise for public health monitoring, policy development, and resource allocation. However, women can and have been arrested for positive drug screens with even preliminary results used to remove children from custody, before rigorous confirmatory testing is completed. Balancing the scientific, medical, public health, legal, and ethical aspects of screening tests for drugs in pregnancy is critical for helping to address this crisis at all levels.
Collapse
Affiliation(s)
- Hayley R Price
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Abby C Collier
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Tricia E Wright
- Department of Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States.,Department of Psychiatry, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| |
Collapse
|
29
|
Detection of in utero ethanol exposure via ethyl glucuronide and ethyl sulfate analysis in umbilical cord and placenta. Forensic Toxicol 2018. [DOI: 10.1007/s11419-018-0439-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
30
|
|
31
|
Paniagua-González L, Jiménez-Morigosa C, Lendoiro E, Concheiro M, Cruz A, López-Rivadulla M, de Castro A. Development and validation of a liquid chromatography-tandem mass spectrometry method for the determination of nicotine and its metabolites in placenta and umbilical cord. Drug Test Anal 2018; 10:1305-1314. [PMID: 29524345 DOI: 10.1002/dta.2381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 11/11/2022]
Abstract
Tobacco exposure during pregnancy is associated with obstetric and fetal complications. We developed and validated a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method to determine nicotine, cotinine, and hydroxycotinine (OH-cotinine) in placenta (PL) and umbilical cord (UC). Specimens were homogenized in water, followed by solid-phase extraction. Chromatographic separation was performed using an Atlantis® HILIC Silica column. Detection was accomplished in electrospray in positive mode. Method validation included: linearity (5 to 1000 ng/g), accuracy (86.9 to 105.2% of target concentration in PL, and 89.1 to 105.0% in UC), imprecision (6.8 to 11.8% in PL, and 7.6 to 12.2% in UC), limits of detection (2 ng/g for cotinine and OH-cotinine, and 5 ng/g for nicotine) and quantification (5 ng/g), selectivity (no endogenous or exogenous interferences), matrix effect (-34.1 to -84.5% in PL, %CV = 9.1-24.0%; -18.9 to -84.7% in UC, %CV = 10.2-23.9%), extraction efficiency (60.7 to 131.5% in PL, and 64.1 to 134.2% in UC), and stability 72 h in the autosampler (<11.5% loss in PL, and < 13% loss in UC). The method was applied to 14 PL and UC specimens from tobacco users during pregnancy. Cotinine (6.8-312.2 ng/g in PL; 6.7-342.3 ng/g in UC) was the predominant analyte, followed by OH-cotinine (
Collapse
Affiliation(s)
- Lucía Paniagua-González
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristian Jiménez-Morigosa
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Elena Lendoiro
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
- School of Pharmacy and Life Sciences, The Robert Gordon University, Aberdeen, UK
| | - Marta Concheiro
- Department of Sciences, John Jay College of Criminal Justice, City University of New York, New York
| | - Angelines Cruz
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel López-Rivadulla
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana de Castro
- Toxicology Service, Institute of Forensic Sciences, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
32
|
Klaman SL, Isaacs K, Leopold A, Perpich J, Hayashi S, Vender J, Campopiano M, Jones HE. Treating Women Who Are Pregnant and Parenting for Opioid Use Disorder and the Concurrent Care of Their Infants and Children: Literature Review to Support National Guidance. J Addict Med 2018; 11:178-190. [PMID: 28406856 PMCID: PMC5457836 DOI: 10.1097/adm.0000000000000308] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/12/2017] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The prevalence of opioid use disorder (OUD) during pregnancy is increasing. Practical recommendations will help providers treat pregnant women with OUD and reduce potentially negative health consequences for mother, fetus, and child. This article summarizes the literature review conducted using the RAND/University of California, Los Angeles Appropriateness Method project completed by the US Department of Health and Human Services Substance Abuse and Mental Health Services Administration to obtain current evidence on treatment approaches for pregnant and parenting women with OUD and their infants and children. METHODS Three separate search methods were employed to identify peer-reviewed journal articles providing evidence on treatment methods for women with OUD who are pregnant or parenting, and for their children. Identified articles were reviewed for inclusion per study guidelines and relevant information was abstracted and summarized. RESULTS Of the 1697 articles identified, 75 were included in the literature review. The perinatal use of medication for addiction treatment (MAT, also known as medication-assisted treatment), either methadone or buprenorphine, within comprehensive treatment is the most accepted clinical practice, as withdrawal or detoxification risks relapse and treatment dropout. Medication increases may be needed with advancing pregnancy, and are not associated with more severe neonatal abstinence syndrome (NAS). Switching medication prenatally is usually not recommended as it can destabilize opioid abstinence. Postnatally, breastfeeding is seen as beneficial for the infant for women who are maintained on a stable dose of opioid agonist medication. Less is known about ideal pain management and postpartum dosing regimens. NAS appears generally less severe following prenatal exposure to buprenorphine versus methadone. Frontline NAS medication treatments include protocol-driven methadone and morphine dosing in the context of nonpharmacological supports. CONCLUSIONS Women with OUD can be treated with methadone or buprenorphine during pregnancy. NAS is an expected and manageable condition. Although research has substantially advanced, opportunities to guide future research to improve maternal and infant outcomes are provided.
Collapse
Affiliation(s)
- Stacey L Klaman
- Department of Maternal and Child Health, University of North Carolina at Chapel Hill Gillings School of Global Public Health, Chapel Hill, NC (SLK); JBS International, Inc., North Bethesda, MD (KI, AL, JP, SH, JV); Center for Substance Abuse Treatment, Substance Abuse and Mental Health Services Administration, US Department of Health and Human Services, Rockville, MD (MC); UNC Horizons, Department of Obstetrics and Gynecology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (HEJ); Departments of Psychiatry and Behavioral Sciences and Obstetrics and Gynecology, School of Medicine, Johns Hopkins University, Baltimore, MD (HEJ)
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Liao MZ, Gao C, Shireman LM, Phillips B, Risler LJ, Neradugomma NK, Choudhari P, Prasad B, Shen DD, Mao Q. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine. Pharmacol Res 2017; 119:61-71. [PMID: 28111265 PMCID: PMC5392442 DOI: 10.1016/j.phrs.2017.01.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 01/11/2023]
Abstract
Norbuprenorphine is the major active metabolite of buprenorphine which is commonly used to treat opiate addiction during pregnancy. Norbuprenorphine produces marked respiratory depression and was 10 times more potent than buprenorphine. Therefore, it is important to understand the mechanism that controls fetal exposure to norbuprenorphine, as exposure to this compound may pose a significant risk to the developing fetus. P-gp/ABCB1 and BCRP/ABCG2 are two major efflux transporters regulating tissue distribution of drugs. Previous studies have shown that norbuprenorphine, but not buprenorphine, is a P-gp substrate. In this study, we systematically examined and compared the roles of P-gp and BCRP in determining maternal brain and fetal distribution of norbuprenorphine using transporter knockout mouse models. We administered 1mg/kg norbuprenorphine by retro-orbital injection to pregnant FVB wild-type, Abcb1a-/-/1b-/-, and Abcb1a-/-/1b-/-/Abcg2-/- mice on gestation day 15. The fetal AUC of norbuprenorphine was ∼64% of the maternal plasma AUC in wild-type mice, suggesting substantial fetal exposure to norbuprenorphine. The maternal plasma AUCs of norbuprenorphine in Abcb1a-/-/1b-/- and Abcb1a-/-/1b-/-/Abcg2-/- mice were ∼2 times greater than that in wild-type mice. Fetal AUCs in Abcb1a-/-/1b-/- and Abcb1a-/-/1b-/-/Abcg2-/- mice were also increased compared to wild-type mice; however, the fetal-to-maternal plasma AUC ratio remained relatively unchanged by the knockout of Abcb1a/1b or Abcb1a/1b/Abcg2. In contrast, the maternal brain-to-maternal plasma AUC ratio in Abcb1a-/-/1b-/- or Abcb1a-/-/1b-/-/Abcg2-/- mice was increased ∼30-fold compared to wild-type mice. Protein quantification by LC-MS/MS proteomics revealed significantly higher amounts of P-gp protein in the wild-type mice brain than that in the placenta. These results indicate that fetal exposure to norbuprenorphine is substantial and that P-gp has a minor impact on fetal exposure to norbuprenorphine, but plays a significant role in restricting its brain distribution. The differential impacts of P-gp on norbuprenorphine distribution into the brain and fetus are likely, at least in part, due to the differences in amounts of P-gp protein expressed in the blood-brain and blood-placental barriers. BCRP is not as important as P-gp in determining both the systemic and tissue exposure to norbuprenorphine. Finally, fetal AUCs of the metabolite norbuprenorphine-β-d-glucuronide were 3-7 times greater than maternal plasma AUCs, while the maternal brain AUCs were <50% of maternal plasma AUCs, suggesting that a reversible pool of conjugated metabolite in the fetus may contribute to the high fetal exposure to norbuprenorphine.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/analysis
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Blood-Brain Barrier/metabolism
- Brain/metabolism
- Buprenorphine/administration & dosage
- Buprenorphine/analogs & derivatives
- Buprenorphine/metabolism
- Buprenorphine/pharmacokinetics
- Female
- Gene Knockout Techniques
- Maternal Exposure
- Maternal-Fetal Exchange
- Mice
- Mice, Knockout
- Narcotic Antagonists/administration & dosage
- Narcotic Antagonists/metabolism
- Narcotic Antagonists/pharmacokinetics
- Pregnancy
- Tissue Distribution
Collapse
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Laura M Shireman
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Brian Phillips
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Linda J Risler
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Naveen K Neradugomma
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Prachi Choudhari
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
34
|
Serra AE, Lemon LS, Mokhtari NB, Parks WT, Catov JM, Venkataramanan R, Caritis SN. Delayed villous maturation in term placentas exposed to opioid maintenance therapy: a retrospective cohort study. Am J Obstet Gynecol 2017; 216:418.e1-418.e5. [PMID: 28024988 DOI: 10.1016/j.ajog.2016.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Accepted: 12/16/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Opioid use disorder among pregnant women is associated with adverse perinatal outcomes and is increasing in the United States. The standard of care for pregnant women with opioid use disorder is opioid maintenance therapy including either methadone or buprenorphine, which can be initiated at any time during pregnancy. These medications are known to cross the placenta but their placental and fetal effects have not been well characterized. Delayed villous maturation, a placental finding associated with stillbirth, was observed in placentas exposed to opioid maintenance therapy. Given the association of delayed villous maturation with stillbirth, and the possible relationship between opioid maintenance therapy and delayed villous maturation, this study was undertaken to explore the association between opioid maintenance therapy and this placental finding. Delayed villous maturation was not previously reported in placentas exposed to opioids or opioid maintenance therapy. OBJECTIVE This study sought to compare risk of delayed villous maturation in term placentas exposed and unexposed to opioid maintenance therapy with buprenorphine or methadone. STUDY DESIGN This was a retrospective cohort study conducted between 2010 through 2012 at Magee-Womens Hospital comparing delayed villous maturation in placentas of women with opioid use disorder exposed to either buprenorphine (n = 86) or methadone (n = 268) versus women without opioid use disorder (n = 978). Potential covariates were assessed in univariate analyses with none significantly associated with delayed villous maturation. The final model used conditional logistic regression adjusting for smoking status alone. RESULTS Among women without opioid use disorder (and therefore not exposed to opioid maintenance therapy), delayed villous maturation was identified in 5.7% of placentas while the prevalence among women treated with buprenorphine or methadone was 8.1% and 10.8%. Overall, the crude odds of being diagnosed with delayed villous maturation were significantly greater in those exposed to opioid maintenance therapy compared to those not exposed (odds ratio, 1.86; 95% confidence interval, 1.20-2.89). When considered separately, women treated with methadone had significantly greater odds of having a placenta with delayed villous maturation than women without exposure to opioid maintenance therapy (odds ratio, 2.00; 95% confidence interval, 1.52-3.20). Women treated with buprenorphine did not have significantly greater odds of this placental diagnosis when compared to the women unexposed to opioid maintenance therapy (odds ratio, 1.46; 95% confidence interval, 0.64-3.31). Results were similar after accounting for smoking. CONCLUSION Delayed villous maturation was more common in the placentas of women exposed to opioid maintenance therapy. Further studies are required to characterize rates and extent of delayed villous maturation in the general population as well as to differentiate between possible effects of opioid exposure (eg, heroin, illicit use of prescription opioids) vs those of opioid maintenance therapy (buprenorphine and methadone).
Collapse
|
35
|
Abstract
Drug testing, when carefully collected and thoughtfully interpreted, offers a critical adjunct to clinical care and substance use treatment. However, because test results can be misleading if not interpreted in the correct clinical context, clinicians should always conduct a careful interview with adolescent patients to understand what testing is likely to show and then use testing to validate or refute their expectations. Because of the ease with which samples can be tampered, providers should also carefully reflect on their own collection protocols and sample validation procedures to ensure optimal accuracy."
Collapse
Affiliation(s)
- Scott E. Hadland
- Boston Children’s Hospital, Division of Adolescent / Young Adult Medicine, Boston Children’s Hospital, Division of Developmental Medicine, Department of Medicine, 300 Longwood Avenue, Boston, MA, USA, 02115,Harvard Medical School, Department of Pediatrics, 25 Shattuck St., Boston, MA, USA, 02115
| | - Sharon Levy
- Department of Medicine, 300 Longwood Avenue, Boston, MA, USA, 02115,Harvard Medical School, Department of Pediatrics, 25 Shattuck St., Boston, MA, USA, 02115
| |
Collapse
|
36
|
Shah D, Brown S, Hagemeier N, Zheng S, Kyle A, Pryor J, Dankhara N, Singh P. Predictors of neonatal abstinence syndrome in buprenorphine exposed newborn: can cord blood buprenorphine metabolite levels help? SPRINGERPLUS 2016; 5:854. [PMID: 27386303 PMCID: PMC4919189 DOI: 10.1186/s40064-016-2576-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 06/15/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Buprenorphine is a semi-synthetic opioid used for the treatment of opioid dependence. Opioid use, including buprenorphine, has been increasing in recent years, in the general population and in pregnant women. Consequently, there has been a rise in frequency of neonatal abstinence syndrome (NAS), associated with buprenorphine use during pregnancy. The purpose of this study was to investigate correlations between buprenorphine and buprenorphine-metabolite concentrations in cord blood and onset of NAS in buprenorphine exposed newborns. METHODS Nineteen (19) newborns who met inclusion criteria were followed after birth until discharge in a double-blind non-intervention study, after maternal consent. Cord blood and tissue samples were collected and analyzed by liquid chromatography-mass spectrometry (LC-MS) for buprenorphine and metabolites. Simple and multiple logistic regressions were used to examine relationships between buprenorphine and buprenorphine metabolite concentrations in cord blood and onset of NAS, need for morphine therapy, and length of stay. RESULTS Each increase in 5 ng/ml level of norbuprenorphine in cord blood increases odds of requiring treatment by morphine 2.5 times. Each increase in 5 ng/ml of buprenorphine-glucuronide decreases odds of receiving morphine by 57.7 %. Along with concentration of buprenorphine metabolites, birth weight and gestational age also play important roles, but not maternal buprenorphine dose. CONCLUSIONS LC-MS analysis of cord blood concentrations of buprenorphine and metabolites is an effective way to examine drug and metabolite levels in the infant at birth. Cord blood concentrations of the active norbuprenorphine metabolite and the inactive buprenorphine-glucuronide metabolite show promise in predicting necessity of treatment of NAS. These finding have implications in improving patient care and reducing healthcare costs if confirmed in a larger sample.
Collapse
Affiliation(s)
- Darshan Shah
- />Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN USA
| | - Stacy Brown
- />Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN USA
| | - Nick Hagemeier
- />Department of Pharmacy Practice, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN USA
| | - Shimin Zheng
- />College of Public Health, East Tennessee State University, Johnson City, TN USA
| | - Amy Kyle
- />Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN USA
| | - Jason Pryor
- />Neonatal Perinatal Medicine, Vanderbilt University Medical Center/Monroe Carell Children’s Hospital, 2200 Children’s Way, Nashville, TN 37232 USA
| | - Nilesh Dankhara
- />Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN USA
| | - Piyuesh Singh
- />Department of Pediatrics, Quillen College of Medicine, East Tennessee State University, Johnson City, TN USA
| |
Collapse
|
37
|
Abstract
Consumption of drugs of abuse, tobacco and alcohol throughout pregnancy is a serious public health problem and results in an important economic cost to the health system. Drug and/or metabolites determination in biological matrices from mother and newborn is an objective measure of in utero drug exposure. We reviewed methods published for the determination of in utero drug exposure from 2007 to 2014, with special focus on meconium, placenta, umbilical cord and newborn hair. Accurate bioanalytical procedures are essential to obtain high-quality data to perform interventions and to establish correlations between analytical measures and clinical outcomes. We included a brief overview of clinical implications of in utero drug exposure to better understand the importance of this serious health issue.
Collapse
|
38
|
Goodman DJ, Milliken CU, Theiler RN, Nordstrom BR, Akerman SC. A Multidisciplinary Approach to the Treatment of Co-occurring Opioid Use Disorder and Posttraumatic Stress Disorder in Pregnancy: A Case Report. J Dual Diagn 2015; 11:248-57. [PMID: 26457976 PMCID: PMC4762276 DOI: 10.1080/15504263.2015.1104484] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Perinatal opioid use disorders negatively impact maternal and neonatal outcomes and are a public health problem of increasing severity. More than half of women with a substance use disorder have a history of posttraumatic stress disorder that, if not adequately addressed, can impede substance use disorder treatment. This case report describes complexities in the treatment of a pregnant woman with opioid use disorder and posttraumatic stress disorder and reviews the psychotherapeutic and pharmacologic approaches available to treat these co-occurring disorders in pregnancy. This case demonstrates the importance of early screening and intervention for co-occurring posttraumatic stress disorder in pregnant women who use substances in a closely coordinated, multidisciplinary approach to improve outcomes for women and their infants.
Collapse
Affiliation(s)
- Daisy J Goodman
- a Department of Obstetrics and Gynecology , Dartmouth-Hitchcock Medical Center and Geisel School of Medicine , Lebanon , New Hampshire , USA
| | - Catherine U Milliken
- b Department of Psychiatry , Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire , USA
| | - Regan N Theiler
- a Department of Obstetrics and Gynecology , Dartmouth-Hitchcock Medical Center and Geisel School of Medicine , Lebanon , New Hampshire , USA
| | - Benjamin R Nordstrom
- b Department of Psychiatry , Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire , USA
| | - Sarah C Akerman
- b Department of Psychiatry , Geisel School of Medicine at Dartmouth , Lebanon , New Hampshire , USA
| |
Collapse
|
39
|
Alternative matrices for cocaine, heroin, and methadone in utero drug exposure detection. Ther Drug Monit 2014; 35:502-9. [PMID: 23851907 DOI: 10.1097/ftd.0b013e31828a6148] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Drug determination in biological matrices from the mother and the newborn is an objective measure of maternal and fetal drug exposure. The aim of this study was to compare maternal hair, meconium, umbilical cord, and placenta for detecting in utero drug exposure to cocaine, opiates, methadone, and amphetamines. METHOD Maternal hair, meconium, umbilical cord, and placenta were collected from 175 mother-newborn dyads. Maternal hair (segmented in trimesters) and meconium specimens were analyzed for cocaine, opiates, methadone, and amphetamines. If either maternal hair or meconium tested positive, umbilical cord and placenta were analyzed. Analyses were performed by liquid chromatography tandem mass spectrometry. RESULTS In hair, 24 participants tested positive; 21 for cocaine [cocaine 20-50,605, benzoylecgonine (BE) 17-46,668 pg/mg], 7 for methadone (76-26,845 pg/mg), 2 for opiates (morphine 298-2398 pg/mg, codeine 65-914 pg/mg, 6-acetylmorphine 1635-15,657 pg/mg), and 1 for amphetamines (amphetamine 1990 pg/mg, 3,4-methylenedioxyamphetamine 30 pg/mg, 3,4-methylenedioxymethamphetamine 294 pg/mg). In meconium, 6 were positive; 5 for methadone [methadone 88-3752, 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) 642-25,179 ng/g], 3 for cocaine (cocaine 7, BE 79, hydroxybenzoylecgonine 5-135, ecgonine-methyl ester 2-56 ng/g), and 2 for opiates (morphine 152-1025, morphine-3-glucuronide 22-23, codeine 4-34 ng/g). Placenta and umbilical cord were positive in 5 and 6 cases, respectively; 5 for methadone in placenta (methadone 7-543, EDDP 10-51 ng/g) and cord (methadone 3-183, EDDP 2-109 ng/g); 1 for cocaine in placenta (cocaine 7, BE 2 ng/g) and cord (BE 6 ng/g); and 1 for opiates in placenta (morphine 6, morphine-3-glucuronide 48 ng/g), and 2 in cord (morphine 2, morphine-3-glucuronide 15-38, morphine-6-glucuronide 5 ng/g). Meconium, placenta, and umbilical cord only tested positive if hair concentrations were greater than Society of Hair Testing cutoffs. CONCLUSIONS Maternal hair is the most sensitive specimen to detect drug consumption during pregnancy. Placenta and umbilical cord could be alternatives to meconium for detecting high in utero drug exposure.
Collapse
|
40
|
Patel P, Abdel-Latif ME, Hazelton B, Wodak A, Chen J, Emsley F, Feller JM, Lui K, Oei JL. Perinatal outcomes of Australian buprenorphine-exposed mothers and their newborn infants. J Paediatr Child Health 2013; 49:746-53. [PMID: 23745982 DOI: 10.1111/jpc.12264] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2013] [Indexed: 11/30/2022]
Abstract
AIM To determine the short-term outcomes of Australian buprenorphine-exposed mother/infant dyads. METHODS Retrospective record review of drug-exposed mothers and infants in Australia. Groups were based on drug exposure: buprenorphine (55, 3.8%), non-buprenorphine opiates (O, 686, 48.6%) and non-opiates (NO, 671, 47.5%). RESULTS More than 30% of buprenorphine mothers continued to use heroin (21, 38%) and benzodiazepines (16, 29%). They were more likely to have child at risk concerns (29, 52.7%, P = 0.019) and have previous children placed in out-of-home care (9, 16.3%, P = 049). Buprenorphine babies were less likely to be preterm (16% vs. 25% (O), P = 0.001 and 23% (NO), P = 0.004) and had higher birthweights (median: 3165 g vs. 2842.5 g (O), P < 0.001 and 2900 g (NO), P = 0.004). Buprenorphine and non-buprenorphine opioid babies had similar maximum Finnegan scores (median 10 vs. 11(O), P = 0.144). The number of babies needing abstinence treatment (45% vs. 51% (O), P = 0.411) and length of hospital stay (median days 9 vs. 11(O), P = 0.067) were similar, but buprenorphine infants required lower maximum morphine doses (mg/kg/day) (median 0.4 mg vs. 0.5 mg (O), P = 0.009). CONCLUSIONS Short-term medical outcomes of infants of buprenorphine-using mothers are similar to those of non-buprenorphine opiate-using mothers, but interpretation of these results is confounded by the high rates of polydrug exposure in the buprenorphine group. This and other social concerns noted in buprenorphine mothers and infants warrant further study.
Collapse
Affiliation(s)
- Pankaj Patel
- The Department of Newborn Care, Royal Hospital for Women, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Pregnancy in opioid users poses a number of problems to treating physicians. Most guidelines recommend maintenance treatment to manage opioid addiction in pregnancy, with methadone being the gold standard. More recently, buprenorphine has been discussed as an alternate medication. The use and efficacy of buprenorphine in pregnancy is still controversial. This article reviews the current database on the basis of a detailed and critical literature search performed in MEDLINE (206 counts). Most of the relevant studies (randomised clinical trials and one national cohort sample) were published in the last 2 years and mainly compared buprenorphine with methadone. Some studies are related to maternal outcomes, others to foetal, neonatal or older child outcomes. With respect to maternal outcomes, most studies suggest that buprenorphine has similar effects to methadone. Very few data from small studies discuss an effect of buprenorphine on neurodevelopment of the foetus. Neonatal abstinence syndrome is common in infants of both buprenorphine- and methadone-maintained mothers. As regards neonatal outcomes, buprenorphine has the same clinical outcome as methadone, although some newer studies suggest that it causes fewer withdrawal symptoms. Since hardly any studies have investigated the combination of buprenorphine with naloxone (which has been suggested to possibly have teratogenic effects) in pregnant women, a switch to buprenorphine monotherapy is recommended in women who become pregnant while receiving the combination product. These novel findings indicate that buprenorphine is emerging as a first-line treatment for pregnant opioid users.
Collapse
|
42
|
Simultaneous determination of opiates, methadone, amphetamines, cocaine, and metabolites in human placenta and umbilical cord by LC-MS/MS. Anal Bioanal Chem 2013; 405:4295-305. [DOI: 10.1007/s00216-013-6784-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/19/2012] [Accepted: 01/24/2013] [Indexed: 10/27/2022]
|
43
|
Launiainen T, Nupponen I, Halmesmäki E, Ojanperä I. Meconium drug testing reveals maternal misuse of medicinal opioids among addicted mothers. Drug Test Anal 2013; 5:529-33. [DOI: 10.1002/dta.1459] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 12/21/2012] [Accepted: 12/21/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Terhi Launiainen
- Hjelt Institute, Department of Forensic Medicine; University of Helsinki; Helsinki; Finland
| | - Irmeli Nupponen
- Hospital for Children and Adolescents; Helsinki University Central Hospital; Helsinki; Finland
| | - Erja Halmesmäki
- Hospital for Children and Adolescents; Helsinki University Central Hospital; Helsinki; Finland
| | - Ilkka Ojanperä
- Hjelt Institute, Department of Forensic Medicine; University of Helsinki; Helsinki; Finland
| |
Collapse
|
44
|
Jones HE, Arria AM, Baewert A, Heil SH, Kaltenbach K, Martin PR, Coyle MG, Selby P, Stine SM, Fischer G. Buprenorphine treatment of opioid-dependent pregnant women: a comprehensive review. Addiction 2012; 107 Suppl 1:5-27. [PMID: 23106923 PMCID: PMC4506646 DOI: 10.1111/j.1360-0443.2012.04035.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIMS This paper reviews the published literature regarding outcomes following maternal treatment with buprenorphine in five areas: maternal efficacy, fetal effects, neonatal effects, effects on breast milk and longer-term developmental effects. METHODS Within each outcome area, findings are summarized first for the three randomized clinical trials and then for the 44 non-randomized studies (i.e. prospective studies, case reports and series and retrospective chart reviews), only 28 of which involve independent samples. RESULTS Results indicate that maternal treatment with buprenorphine has comparable efficacy to methadone, although difficulties may exist with current buprenorphine induction methods. The available fetal data suggest buprenorphine results in less physiological suppression of fetal heart rate and movements than methadone. Regarding neonatal effects, perhaps the single definitive conclusion is that prenatal buprenorphine treatment results in a clinically significant less severe neonatal abstinence syndrome (NAS) than treatment with methadone. The limited research suggests that, like methadone, buprenorphine is compatible with breastfeeding. Data available thus far suggest that there are no deleterious effects of in utero buprenorphine exposure on infant development. CONCLUSIONS While buprenorphine produces a less severe neonatal abstinence syndrome than methadone, both methadone and buprenorphine are important parts of a complete comprehensive treatment approach for opioid-dependent pregnant women.
Collapse
Affiliation(s)
- Hendrée E. Jones
- Department of Psychiatry, Johns Hopkins University School of Medicine
| | - Amelia M. Arria
- Center on Young Adult Health and Development, School of Public Health, University of Maryland, College Park
| | - Andjela Baewert
- Department of Psychiatry and Psychotherapy, Medical University Vienna
| | - Sarah H. Heil
- Departments of Psychiatry and Psychology, University of Vermont
| | - Karol Kaltenbach
- Department of Pediatrics, Jefferson Medical College, Thomas Jefferson University
| | | | - Mara G. Coyle
- Department of Pediatrics, The Warren Alpert Medical School of Brown University
| | - Peter Selby
- Departments of Family and Community Medicine, Psychiatry and Public Health Sciences, University of Toronto
| | - Susan M. Stine
- Department of Psychiatry & Behavior Neurosciences, Wayne State University
| | - Gabriele Fischer
- Department of Psychiatry and Psychotherapy, Medical University Vienna
| |
Collapse
|
45
|
Salisbury AL, Coyle MG, O’Grady KE, Heil SH, Martin PR, Stine SM, Kaltenbach K, Weninger M, Jones HE. Fetal assessment before and after dosing with buprenorphine or methadone. Addiction 2012; 107 Suppl 1:36-44. [PMID: 23106925 PMCID: PMC4277183 DOI: 10.1111/j.1360-0443.2012.04037.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIM To determine pre- and post-dosing effects of prenatal methadone compared to buprenorphine on fetal wellbeing. DESIGN A secondary analysis of data derived from the Maternal Opioid Treatment: Human Experimental Research (MOTHER) study, a double-blind, double-dummy, randomized clinical trial. SETTING Six United States sites and one European site that provided comprehensive opioid-dependence treatment to pregnant women. PARTICIPANTS Eighty-one of the 131 opioid-dependent pregnant women completing the MOTHER clinical trial, assessed between 31 and 33 weeks of gestation. MEASUREMENTS Two fetal assessments were conducted, once before and once after study medication dosing. Measures included mean fetal heart rate (FHR), number of FHR accelerations, FHR reactivity in the fetal non-stress test (NST) and biophysical profile (BPP) score. FINDINGS Significant group differences were found for number of FHR accelerations, non-reactive NST and BPP scores (all Ps < 0.05). There were no significant group differences before time of dosing. Significant decreases (all Ps < 0.05) occurred from pre- to post-dose assessment for mean FHR, FHR accelerations, reactive NST and fetal movement. The decrease in accelerations and reactive NST were significant only for fetuses in the methadone group, and this resulted in a significantly lower likelihood of a reactive NST compared to fetuses in the buprenorphine group. CONCLUSION Buprenorphine compared with methadone appears to result in less suppression of mean fetal heart rate, fetal heart rate reactivity and the biophysical profile score after medication dosing and these findings provide support for the relative safety of buprenorphine when fetal indices are considered as part of the complete risk-benefit ratio.
Collapse
Affiliation(s)
- Amy L. Salisbury
- Department of Pediatrics and Department of Psychiatry and Human Behavior, The Warren Alpert Medical School of Brown University
| | - Mara G. Coyle
- Department of Pediatrics, The Warren Alpert Medical School of Brown University
| | | | - Sarah H. Heil
- Departments of Psychiatry and Psychology, University of Vermont
| | - Peter R. Martin
- Departments of Psychiatry and Pharmacology and Addiction Center, Vanderbilt University School of Medicine
| | - Susan M. Stine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine
| | - Karol Kaltenbach
- Department of Pediatrics, Jefferson Medical College, Thomas Jefferson University
| | - Manfred Weninger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Medical University of Vienna
| | - Hendrée E. Jones
- Department of Psychiatry and Behavioral Sciences and Department of Obstetrics and Gynecology, Johns Hopkins University School of Medicine, Substance Abuse Treatment Evaluations and Interventions Program, RTI International
| |
Collapse
|
46
|
Falcon M, Pichini S, Joya J, Pujadas M, Sanchez A, Vall O, Algar OG, Luna A, de la Torre R, Rotolo M, Pellegrini M. Maternal hair testing for the assessment of fetal exposure to drug of abuse during early pregnancy: Comparison with testing in placental and fetal remains. Forensic Sci Int 2012; 218:92-6. [DOI: 10.1016/j.forsciint.2011.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 06/14/2011] [Indexed: 12/31/2022]
|
47
|
Bartu AE, Ilett KF, Hackett LP, Doherty DA, Hamilton D. Buprenorphine exposure in infants of opioid-dependent mothers at birth. Aust N Z J Obstet Gynaecol 2012; 52:342-7. [PMID: 22428721 DOI: 10.1111/j.1479-828x.2012.01424.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 02/05/2012] [Indexed: 11/29/2022]
Affiliation(s)
- Anne E. Bartu
- Faculty of Health Sciences; School of Nursing and Midwifery; Curtin University of Technology; Australia
| | - Kenneth F. Ilett
- Pharmacology, Pharmacy and Anaesthesiology Unit; School of Medicine and Pharmacology; University of Western Australia; Australia
- Clinical Pharmacology and Toxicology Laboratory; Path West Laboratory Medicine; Australia
| | - L. Peter Hackett
- Clinical Pharmacology and Toxicology Laboratory; Path West Laboratory Medicine; Australia
| | - Dorota A. Doherty
- School of Women's and Infants' Health; University of Western Australia; Australia
| | - Dale Hamilton
- King Edward Memorial Hospital; Women and Newborn Health Services; Western Australia Australia
| |
Collapse
|
48
|
Hendrickson RG, McKeown NJ. Is maternal opioid use hazardous to breast-fed infants? Clin Toxicol (Phila) 2011; 50:1-14. [DOI: 10.3109/15563650.2011.635147] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Concheiro M, Jones HE, Johnson RE, Choo R, Huestis MA. Preliminary buprenorphine sublingual tablet pharmacokinetic data in plasma, oral fluid, and sweat during treatment of opioid-dependent pregnant women. Ther Drug Monit 2011; 33:619-26. [PMID: 21860340 PMCID: PMC3178674 DOI: 10.1097/ftd.0b013e318228bb2a] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Buprenorphine is currently under investigation as a pharmacotherapy to treat pregnant women for opioid dependence. This research evaluates buprenorphine (BUP), norbuprenophine (NBUP), buprenorphine-glucuronide (BUP-Gluc), and norbuprenorphine-glucuronide (NBUP-Gluc) pharmacokinetics after high-dose (14-20 mg) BUP sublingual tablet administration in three opioid-dependent pregnant women. METHODS Oral fluid and sweat specimens were collected in addition to plasma specimens for 24 hours during gestation weeks 28 or 29 and 34, and 2 months after delivery. Time to maximum concentration was not affected by pregnancy; however, BUP and NBUP maximum concentration and area under the curve at 0 to 24 hours tended to be lower during pregnancy compared with postpartum levels. RESULTS Statistically significant but weak positive correlations were found for BUP plasma and OF concentrations and BUP/NBUP ratios in plasma and oral fluid. Statistically significant negative correlations were observed for times of specimen collection and BUP and NBUP oral fluid/plasma ratios. BUP-Gluc and NBUP-Gluc were detected in only 5% of oral fluid specimens. In sweat, BUP and NBUP were detected in only four of 25 (12 or 24 hours) specimens in low concentrations (less than 2.4 ng/patch). CONCLUSION These preliminary data describe BUP and metabolite pharmacokinetics in pregnant women and suggest that, like methadone, upward dose adjustments may be needed with advancing gestation.
Collapse
Affiliation(s)
- Marta Concheiro
- Servicio de Toxicología Forense, Instituto de Ciencias Forenses, Universidad de Santiago de Compostela, Spain
- Chemistry and Drug Metabolism, IRP, National Institute on Drug Abuse, NIH, Baltimore, MD
| | - Hendreé E. Jones
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rolley E. Johnson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
- Reckitt Benckiser Pharmaceuticals, Inc., Richmond, VA
| | - Robin Choo
- Department of Biology, University of Pittsburgh, Titusville, PA
| | - Marilyn A. Huestis
- Chemistry and Drug Metabolism, IRP, National Institute on Drug Abuse, NIH, Baltimore, MD
| |
Collapse
|