1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      P-glycoprotein efflux transporter: a key to pharmacokinetic modeling for methadone clearance in fetuses

      discussion
      1 , * , , 2 , * ,
      Frontiers in Pharmacology
      Frontiers Media S.A.
      methadone, opioids, PBPK, P-glycoprotein, model

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background The situation of opioid abuse has deteriorated in recent years. In 2019, 49,860 people died from opioid overdoses in the USA alone, a 6-fold increase since 2000 (Ko et al., 2020). Of particular concern is opioid abuse (e.g., morphine, fentanyl) by pregnant women. It was found that 6.6% of women self-reported opioid use during pregnancy in the USA, and 21.2% of them disclosed opioid misuse (Ko et al., 2020). Infants born to drug-abusing mothers are at risk of preterm delivery, poor intrauterine growth, and neonatal abstinence syndrome (NAS). Currently, Methadone Maintenance Treatment (MMT) is the recommended therapy for opioid addicts, including pregnant women (World Health Organization, 2014). However, the pharmacokinetics of methadone is greatly affected by pregnancy. Physiological changes during pregnancy (e.g., increases in total body fluid, blood volume, and body fat) and metabolic enzyme activities (CYP2B6, CYP3A4) lead to faster clearances of methadone in pregnant women (Badhan and Gittins, 2021). Consequently, a higher dosage may be required during pregnancy. Some researchers suggest a staged escalation approach, i.e., starting from the lowest pre-natal dose (30 mg once daily) to the maximum 120, 140, and 180 mg daily dose for trimester 1, 2 and 3, respectively (Badhan and Gittins, 2021). This is significantly higher than a standard dosage (15–60 mg/day) and may expose fetuses and neonates to higher risks of NAS. Indeed, of all infants born to mothers receiving MMT, 40%–90% show signs of NAS. For example, in a retrospective study of 67 women (40 received MMT at 10–20 mg/day), it was found that 43% of the infants of mothers receiving MMT required treatment for NAS (Malpas et al., 1995). Similar results were also reported in (Dryden et al., 2009). However, it was also found that there was no strong correlation between the dosage of methadone use and the severity of NAS (Malpas et al., 1995; Berghella et al., 2003). In a recent pregnant rat model, it was shown that the methadone concentrations in the blood and brain of fetuses were 1.6 and 2.8 times higher than in dams, respectively (Kongstorp et al., 2019) (Figure 1A). The finding highlights the susceptibility of fetuses to maternal methadone exposure. Being lipophilic and with a low molecular weight (309 Da), methadone readily crosses the placenta barrier via passive diffusion. However, the return of methadone from the fetal side to the maternal side (f→m) also requires active transport by the efflux transporter P-glycoprotein (P-gp) (Nanovskaya et al., 2005). P-gp is expressed at the apical membrane of cotyledons, i.e., the maternal side (Gil et al., 2005; Nanovskaya et al., 2005). Therefore, the placenta acts as barrier that retards the backflow of methadone f→m, which causes a higher plasma concentration of methadone at the fetal side than the maternal side (Figure 1A). To assist the clearance of methadone from the fetal side, P-gp acts as the efflux transporter (f→m), on top of the limited passive perfusion. Therefore, an inhibition of P-gp would deteriorate the accumulation of methadone at the fetal side. The phenomenon has been demonstrated in experiments. For example, in an ex vivo placental perfusion experiment, it was shown that inhibition of P-gp led to a 30% increase in methadone in fetal circulation (Nanovskaya et al., 2008). Similar to its role as an efflux transporter in the placenta, P-gp is expressed at the blood-brain barrier (BBB) and acts as an efflux transporter for xenobiotics from brain tissues back to the cerebral flow. Previous studies of P-gp mediated methadone perfusion across the BBB are rarely reported. However, inhibition of P-gp greatly increased the toxicity of another synthetic opioid, fentanyl, in a rat brain model (Yu et al., 2018). The aggregated efflux effects of P-gp on the placental and BBB barriers lead to the higher concentrations in fetal brain not only higher that the maternal brain tissues, but also higher than fetal circulations in rat models (Figure 1A) (Kongstorp et al., 2019). FIGURE 1 (A) The methadone concentrations in the blood and brain of fetuses were 1.6 and 2.8 times higher than in dams, respectively. Figure adopted from Kongstorp et al. (2019); (B) The diagram for a f-m PBPK model for methadone, and the proposed changes to incorporate the P-gp inhibition/induction kinetics. Equations for at least four compartments (the brain, intestine, placenta, and fetus) of the model need to be revised. Based on these observations, it is hypothesized that by inducing P-gp at the placental barrier and BBB, the methadone concentration in the fetal blood and brain can be reduced. Consequently, symptoms of CNS may be alleviated. However, co-administration of a P-gp inducer (e.g., Rifamacin) with methadone also induces metabolic enzymes, e.g., CYP3A4, which can lead to a more rapid clearance of methadone and possible withdrawal syndromes in mothers. In addition, a P-gp inhibitor or inducer acting on the intestine barrier may change the bioavailability of methadone if it is administered orally (Carlos et al., 2002). This is a typical drug-drug interaction (DDI) scenario between methadone, P-gp and cytochrome P450 (CYP) enzymes in a specific population (pregnant women and fetuses), where there is a significant knowledge gap. In this opinion paper, we first provide a brief review of some in vivo, in vitro, and ex vivo models for the role P-gp plays in methadone clearance in fetuses. We then highlight P-gp as a key in fetal-maternal physiologically based pharmacokinetic (PBPK) and ex vivo placenta perfusion models, and propose computational methods for incorporating efflux transporter kinetics in pharmacokinetic models. In vivo animal models As a synthetic opioid agonist, methadone is mainly eliminated via hepatic metabolism by CYP 2B6, and partly by CYP 3A4 (Garrido and Trocóniz, 1999; Kharasch, 2017). Its concentration in the blood has a long half-life (20–35 h), which is crucial to prevent withdrawal symptoms for at least 24 h. In vivo models for fetal exposure to methadone are mainly performed in animals in particular on rats (Gabrielsson et al., 1985; Chen et al., 2015; Kongstorp et al., 2019; Kongstorp et al., 2020). In these models, administering methadone is either via subcutaneous injection (Chen et al., 2015), or through an implanted osmotic minipump to keep constant opioid exposure in dams (Kongstorp et al., 2019). The dose regimen was 10 mg/kg/day, or 1.0 mL/kg of body weight to induce stable blood concentrations of 0.25 ± 0.02 μM methadone in the pregnant rats, which are comparable to the concentrations reported in pregnant women in MMT. However, in vivo data on P-gp mediated fetal exposure to methadone are rare. For example, in a literature review of fetal and offspring exposure to opioids in animals and humans, there was no mention of either inhibition or inducing P-gp (Farid et al., 2008). In vitro cell line models Monolayer BeWo cell lines have been used to investigate the permeability of opioids in the placental trophoblast (Nanovskaya et al., 2005; Mortensen et al., 2019). A study investigated the BeWo cell permeability of six opioids (Mortensen et al., 2019), and it was found that the permeability of methadone is lower than oxycodone but higher than morphine and heroin. Another study revealed methadone uptake was increased in the presence of the P-gp inhibitor cyclosporin A (Nanovskaya et al., 2005), reflecting a reduced methadone efflux. In addition, it has been shown that the basolateral to apical transport of known p-glycoprotein substrates (vinblastine, vincristine, and digoxin) is significantly greater than transport to the opposite direction (Ushigome et al., 2000). This is consistent with what has been found in the ex vivo placenta in perfusion experiments (Nanovskaya et al., 2008). Although it is rare, human intestinal cell line Caco-2 was used to investigate transporter-mediated opioid transfer in BBB. The efflux ratio of fentanyl was remarkably reduced when co-incubated with tariquidar, a P-gp inhibitor (Yu et al., 2018). However, the prediction of drug transport through the BBB in vivo may not be accurate since the BBB, and the intestinal musosa are two fundamentally different biologic barriers (Lundquist et al., 2002). Instead, in vitro models of BBB with monolayer of brain capillary endothelial cells were shown to have a good correlation with in vivo rat models (Lundquist et al., 2002). Ex vivo placental models Ex vivo placental perfusion is considered the “golden standard” for examining the transplacental properties of an investigational drug. In the experiment setup, a single unit of cotyledon is cannulated and connected to perfusates at the maternal and fetal sides separately (Nanovskaya et al., 2008) (Malek et al., 2009) (Kurosawa et al., 2020). In one such study (Nanovskaya et al., 2008), methadone concentrations were measured from the perfusate reservoirs and at sampling ports. It was found that in the maternal to fetal (m→f) perfusion route, there was a biphasic decrease in methadone concentration, i.e., a sharp drop in the first 60 min followed by a plateau in the remaining 180 min. In contrast, the f→m transperfusion showed a gradual decrease of methadone in the fetal artery. The opiate was retained by the placental tissue from both the m→f and the f→m routes. It is worth pointing out that since placentas are usually collected postpartum, the ex vivo perfusion results only reflect the placental physiology at term, but not at the early stages of gestation. For example, the P-gp expression is lower at the apical membrane in trimester 3 than in other trimesters, which leads to a weaker efflux transport, and consequently a higher methadone accumulation in trimester 3 (Gil et al., 2005). This is unfortunate because the last stage of gestation is also the most crucial time for lowering fetal exposure to opioids and reducing the risk of NAS. In silico models PBPK models are computational tools that quantify the time course of drug concentration in various organs and tissues, represented by compartments (Figure 1B). Fetal-maternal (f-m) PBPK models add compartments of the placenta and fetal organs into a standard model. f-m PBPK models have been developed for many drugs, including methadone (Gabrielsson et al., 1985; Ke et al., 2014). However, to our knowledge, the only pregnant rat model of methadone that has both in vivo experiments and f-m PBPK modelling was developed almost 40 years ago (Gabrielsson et al., 1985). Unfortunately, no DDI mechanism was investigated in that study. In order to incorporate efflux transporter kinetics in the PBPK scheme shown in Figure 1B, four key compartments, namely, the brain, the placenta, the fetus and the intestine need to be revised (refer to Figure 1B). For the brain compartment, the transport-mediated DDI for BBB was implemented in two compartments, i.e., brain tissue and brain vasculature (Ball et al., 2012), where a passive diffusion term and two transport terms (representing active influx and efflux) are used to simulate the transport. A more complex four-compartment model was proposed in (Gaohua et al., 2016), where cranial and spinal cerebrospinal fluid compartments are added. The placenta compartment can be treated in a similar way, i.e., with apical and basolateral membranes in cotyledons, as has been used in an f-m PBPK model for nicotine (Amice et al., 2021). If methadone is administered orally, such as the liquid formulation being practiced in MMT clinics, then the intestine barrier needs to be implemented in the PBPK model. The pharmacokinetics of methadone in this case is different from when it is administered through infusion or intravenous injection (Chen et al., 2015). For instance, for the administration route of methadone, instead of intravenous injection, an oral dosing regimen will also be used for comparison. It is worth noting that adding the fetal liver into the fetus compartment may not be significant for methadone from ontogeny’s perspective, since some of the major metabolic enzymes (CYP2B6, CYP3A4) are still not mature in fetuses, while other enzymes such as CYP3A7 is expressed in the fetal liver that can metabolise methadone (Wolff et al., 2005). Nevertheless, the fetal liver compartment is important for some other opioids including morphine and buprenorphine, because fetal liver enzymes, e.g., SULT1A3 and SULT2A1 that are involved in these opioids’ sulfation are already active. A more thorough investigation of the transport properties of methadone in the placenta would require considering several membranes that separate the maternal and fetal circulations. To date, one of the most detailed mathematical models for ex vivo placenta perfusion is that developed for metformin (Kurosawa et al., 2020) and for morphine (Ho et al., 2022). The significance of the work is that the important data of P-gp mediated methadone transport were published (Nanovskaya et al., 2008), which allows an in silico model to be calibrated. Furthermore, the parameters in in silico models can be tuned per the reduced P-gp expressions as gestation progresses (Nanovskaya et al., 2005). Need for integrating in silico, in vitro, and in vivo models In the above section, we have briefly discussed the current in vivo, in vitro, and in silico models for fetal exposure to methadone, in particular when considering the variable of efflux transporter P-gp. We also described a strategy for incorporating efflux kinetics into PBPK and ex vivo perfusion models. It is clear that none of these models is comprehensive in addressing the multiple facets of the methadone clearance mechanism in pregnant women and fetuses. Still, these models need to provide evidence-based methodology change for the current MMT practice, where clinicians rely on clinical symptoms (withdrawal or overmedication) when prescribing methadone. Efforts need to be made to fill the gaps and challenges. For example, extrapolation of in vivo placenta transfer from animal models to humans is challenging due to the anatomical and functional specificity of the placenta among different species (Myllynen and Vähäkangas, 2013). Similarly, in vitro cell line models may fall short of the prediction of in vivo drug transport (Lundquist et al., 2002). Ex vivo placenta perfusion models can only predict opioid transport properties at term, yet such properties are clearly altered by varying P-gp expression during pregnancy. While in silico models are cost-effective, they are prone to mathematical maneuver or parameter fitting when lacking data (Ho et al., 2022). Nevertheless, there is a more pressing need than ever to integrate the data and know-how gained from multiscale, multidisciplinary models in the era of the “opioid epidemic” and its treatment.

          Related collections

          Most cited references30

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Vital Signs: Prescription Opioid Pain Reliever Use During Pregnancy — 34 U.S. Jurisdictions, 2019

          Abstract Background: Prescription opioid use during pregnancy has been associated with poor outcomes for mothers and infants. Studies using administrative data have estimated that 14%–22% of women filled a prescription for opioids during pregnancy; however, data on self-reported prescription opioid use during pregnancy are limited. Methods: CDC analyzed 2019 data from the Pregnancy Risk Assessment Monitoring System (PRAMS) survey in 32 jurisdictions and maternal and infant health surveys in two additional jurisdictions not participating in PRAMS to estimate self-reported prescription opioid pain reliever (prescription opioid) use during pregnancy overall and by maternal characteristics among women with a recent live birth. This study describes source of prescription opioids, reasons for use, want or need to cut down or stop use, and receipt of health care provider counseling on how use during pregnancy can affect an infant. Results: An estimated 6.6% of respondents reported prescription opioid use during pregnancy. Among these women, 21.2% reported misuse (a source other than a health care provider or a reason for use other than pain), 27.1% indicated wanting or needing to cut down or stop using, and 68.1% received counseling from a provider on how prescription opioid use during pregnancy could affect an infant. Conclusions and Implications for Public Health Practice: Among respondents reporting opioid use during pregnancy, most indicated receiving prescription opioids from a health care provider and using for pain reasons; however, answers from one in five women indicated misuse. Improved screening for opioid misuse and treatment of opioid use disorder in pregnant patients might prevent adverse outcomes. Implementation of public health strategies (e.g., improving state prescription drug monitoring program use and enhancing provider training) can support delivery of evidence-based care for pregnant women. Introduction During 2017–2018, 42.5% of opioid-related overdose deaths among women in the United States involved a prescription opioid ( 1 ). Long-term use of prescription opioids is associated with increased risk for misuse (i.e., use in larger amounts, higher frequency, longer duration, or for a different reason than that directed by a prescribing physician) ( 2 ), opioid use disorder, and overdose ( 3 , 4 ). According to commercial insurance ( 5 ) and Medicaid ( 6 ) claims for reimbursement of pharmacy dispensing, an estimated 14%–22% of women filled at least one opioid prescription during pregnancy ( 5 , 6 ). Opioid use during pregnancy has been associated with poor infant outcomes, such as neonatal opioid withdrawal syndrome ( 7 ), preterm birth, poor fetal growth, and stillbirth ( 8 ). PRAMS* and two additional jurisdictions’ maternal and infant health surveys conducted during 2019 were used to describe population-based, self-reported estimates of prescription opioid pain reliever (prescription opioid) use during pregnancy. Methods PRAMS is a jurisdiction-specific and population-based surveillance system designed to monitor self-reported behaviors and experiences before, during, and shortly after pregnancy among women with a live birth in the preceding 2–6 months. Detailed PRAMS methodology is published elsewhere ( 9 ). Supplementary questions on prescription opioid use during pregnancy were asked in 32 jurisdictions participating in PRAMS and on maternal and infant health surveys in two jurisdictions that do not participate in PRAMS. † Data were weighted to adjust for sample design and nonresponse, representing the total population of women with a live birth in each jurisdiction during an approximately 4-month § or 5-month ¶ period in 2019. Women were asked, “During your most recent pregnancy, did you use any of the following prescription pain relievers?” Use of prescription opioid pain relievers (prescription opioids) during pregnancy was indicated by selection of any of the following: hydrocodone, codeine, oxycodone, tramadol, hydromorphone or meperidine, oxymorphone, morphine, or fentanyl.** Women who self-reported use during pregnancy were asked to check all that apply to additional questions describing the prescription opioid source and reasons for use. †† Qualitative thematic coding was used to recode “other” written-in text responses into existing and new categories, where possible. §§ Remaining responses were retained as “other/undetermined.” Prescription opioid sources were categorized as health care and non–health care provider (based on the responses “I had pain relievers left over from an old prescription,” “friend or family member gave them to me,” or “I got the pain relievers without a prescription some other way”). Reasons for use were categorized as pain and any reason other than pain (based on the responses “to relax or relieve tension or stress,” “to help me with feelings or emotions,” “to help me sleep,” “to feel good or get high,” or “because I was ‘hooked’ or I had to have them”). Misuse was defined as getting opioids from any source other than a health care provider or using for any reason other than pain. Respondents were also asked about their desire to cut down or stop use (“During your most recent pregnancy, did you want or need to cut down or stop using prescription pain relievers?”) and whether they received provider counseling (“At any time during your most recent pregnancy, did a doctor, nurse, or other health care worker talk with you about how using prescription pain relievers during pregnancy could affect a baby?”). Prevalence of prescription opioid use during pregnancy was estimated overall and by maternal characteristics. Maternal age, race/ethnicity, education, trimester of entry into prenatal care, health insurance at delivery, and number of previous live births were derived from birth certificate data. Self-reported cigarette use during the last 3 months of pregnancy and depression during pregnancy were obtained from the surveys. Among women reporting prescription opioid use during pregnancy, estimates were generated for source, reasons for use, want or need to cut down or stop use, and receipt of health care provider counseling on how use during pregnancy could affect an infant. Prevalence of receipt of health care provider counseling was estimated by maternal characteristics. In addition, the percentage of women who wanted or needed to cut down or stop using was estimated among those who reported misuse as defined in this study and those who did not. Chi-squared tests were used to assess the differential distribution of prescription opioid use during pregnancy and receipt of health care provider counseling by maternal characteristics, as well as the want or need to cut down or stop use by misuse classification. Weighted prevalence estimates and 95% confidence intervals (CIs) were calculated using SUDAAN (version 11.0; RTI International). Results In 2019, among 21,488 respondents, 20,643 (96.1%) provided information regarding prescription opioid use during their most recent pregnancy. Among these women, 1,405 (6.6%) reported prescription opioid use during pregnancy (Table 1). The prevalence of use was statistically different across the following categories: health insurance at delivery, cigarette smoking during the last 3 months of pregnancy, and depression during pregnancy (p 12 13,415 805 6.1 (5.4–6.9) Trimester of entry into prenatal care First 16,241 1,072 6.2 (5.6–6.9) Second, third, or none 3,124 205 6.3 (4.9–7.9) Health insurance at delivery ¶ Private** 10,653 591 5.2 (4.6–6.0) Medicaid 8,317 712 8.5 (7.5–9.7) Other†† or none 1,068 59 4.4 (2.9–6.5) No. of previous live births None 7,982 504 6.3 (5.4–7.3) One or more 12,508 885 6.7 (6.0–7.5) Smoked cigarettes during last 3 mos of pregnancy ¶ Yes 1,279 192 16.2 (12.7–20.4) No 19,227 1,200 5.9 (5.4–6.5) Depression during pregnancy ¶ , §§ Yes 2,432 295 13.1 (10.7–15.8) No 12,319 730 5.4 (4.8–6.1) Abbreviation: CI = confidence interval. * Unweighted sample size. † Weighted prevalence (expressed as a percentage). § Includes Asian, American Indian, Alaska Native, Native Hawaiian, Pacific Islander, and mixed race/ethnicity. ¶ Indicates chi-squared test p 12 793 515 69.4 (63.9–74.4) Trimester of entry into prenatal care First 1,052 688 70.2 (65.4–74.6) Second, third, or none 200 125 61.6 (49.9–72.2) Health insurance at delivery Private** 582 379 71.6 (65.5–77.0) Medicaid 694 455 67.6 (61.1–73.5) Other†† or none 54 32 57.1 (36.7–75.3)§ No. of previous live births §§ None 494 308 62.0 (54.3–69.2) One or more 863 570 71.6 (66.5–76.2) Smoked cigarettes during last 3 mos of pregnancy Yes 185 107 64.0 (51.4–75.0) No 1,175 770 68.7 (64.0–73.0) Depression during pregnancy ¶¶ Yes 289 195 76.0 (67.2–83.1) No 709 457 65.9 (59.9–71.4) Abbreviation: CI = confidence interval. * Unweighted sample size. † Weighted prevalence (expressed as a percentage). § Denominator is less than <60, so estimate may be unstable. ¶ Includes Asian, American Indian, Alaska Native, Native Hawaiian, Pacific Islander, and mixed race/ethnicity. **Includes Civilian Health and Medical Program of the Department of Uniformed Services and TRICARE. †† Includes Children’s Health Insurance Program and other government programs. §§ Indicates chi-squared test p<0.05. ¶¶ California data not available. Discussion In this population-based sample of women with recent live births in 34 jurisdictions, one in 15 (6.6%) respondents self-reported using prescription opioid pain relievers during pregnancy. This observed prevalence of use during pregnancy in 2019 is lower than estimates of prescription opioid fills from administrative data (e.g., insurance claims) in previous years ( 5 , 6 ), which do not necessarily correlate with use. Higher use of prescription opioids among women who reported smoking cigarettes or had depression during pregnancy are consistent with findings from studies analyzing administrative Medicaid data ( 7 ). In this study, an estimated one in five women using prescription opioids during pregnancy indicated misuse. In addition, more than one in four (27.1%) women with prescription opioid use indicated wanting or needing to reduce or stop their use, potentially because of concerns about the effect of medication on their infant, possible opioid dependence, or opioid use disorder. Among women reporting prescription opioid use, nearly one in three (31.9%) reported not receiving provider counseling on the effects of prescription opioid use on an infant. Clinical guidance addresses opioid prescribing and tapering during pregnancy, the risks to the mother and infant, and screening and treatment for opioid dependence and opioid use disorder ( 3 , 10 ). CDC and the American College of Obstetricians and Gynecologists (ACOG) recommend that clinicians and patients discuss and carefully weigh risks and benefits when considering initiation of opioid therapy for chronic pain during pregnancy ( 3 , 10 ). Opioids, if indicated, should be prescribed only after consideration of alternative pain management therapies ( 3 , 10 ). Risk for physiologic dependence and possibility of an infant developing neonatal opioid withdrawal syndrome should be discussed ( 10 ). Clinicians caring for pregnant women are advised to perform verbal screening to identify and address substance use, misuse, and substance use disorders ( 10 , 11 ). Co-occurring use of other substances (e.g., tobacco) and mental health conditions are more common among pregnant women who are prescribed or misusing prescription opioids than among those who are not ( 7 , 12 ). Recommended screening and, if applicable, treatment and referral for depression, history of trauma, posttraumatic stress disorder, and anxiety should occur ( 10 ). Because of the possible risk for spontaneous abortion and premature labor associated with opioid withdrawal ( 10 ), clinicians are encouraged to consult with other health care providers as necessary if considering tapering opioids during pregnancy ( 3 ). Medications for opioid use disorder, including buprenorphine or methadone, are recommended because of their association with improved maternal outcomes ( 3 , 10 , 13 ). Collaboration between obstetric and neonatal providers is important to diagnose, evaluate, and treat neonatal opioid withdrawal syndrome because it can result from medically indicated opioid prescription use, medication for opioid use disorder, or illicit opioid use ( 3 , 10 ). Effective public health strategies to support the implementation of evidence-based guidelines might include improving state prescription drug monitoring program use ( 14 ), provider training ( 15 ), multidisciplinary state learning communities ( 16 ), quality improvement collaboratives ( 17 ), and consumer awareness ( 18 ). For example, some state perinatal quality collaboratives are implementing the Alliance for Innovation on Maternal Health program’s patient safety obstetric care bundle for pregnant and postpartum women with opioid use disorder to implement protocols for screening and referral to treatment ( 16 , 19 ). The findings in this report are subject to at least five limitations. First, these population-based data are only generalizable to women with a recent live birth in the 34 jurisdictions included in this report. Because of the need to provide data on the opioid crisis among pregnant women, a response rate threshold was not required for jurisdictions to be included in the analyses. This might further affect generalizability because 13 jurisdictions fell below the current PRAMS threshold of 55% ( 9 ). Second, prescription opioid use was self-reported and might be underestimated because of stigma and legal implications. ¶¶ Third, question misinterpretation by respondents is possible. For example, <1% indicated no source or reason for use except for a written-in response regarding use during labor and delivery, even though the initial prompt asked women to not include pain relievers used during labor and delivery. Fourth, not all available misuse indicators (e.g., use for longer time than prescribed) were assessed. Finally, the opioid supplement questions do not reflect current diagnostic criteria and cannot be used to estimate the prevalence of opioid use disorder ( 20 ). Opioid prescribing consistent with clinical practice guidelines can ensure that patients, particularly those who are pregnant, have access to safer, more effective chronic pain treatment and reduce the number of persons at risk for opioid misuse, opioid use disorder, and overdose. Implementation of public health strategies can complement these efforts to improve the health of mothers and infants. The PRAMS surveillance system can be used to identify opportunities for providers, health systems, and jurisdictions to better support pregnant and postpartum women and their families. Summary What is already known about this topic? Data on self-reported prescription opioid use during pregnancy are limited. What is added by this report? Analysis of 2019 survey data found that 6.6% of women reported prescription opioid use during pregnancy. Among these women, 21.2% reported misuse (a source other than a health care provider or a reason for use other than pain), 27.1% wanted or needed to cut down or stop using, and 31.9% reported not receiving provider counseling about how use could affect an infant. What are the implications for public health practice? Obstetric providers should discuss risks and benefits of opioid therapy for chronic pain during pregnancy, screen all pregnant women for substance use, misuse, and use disorders, including those involving prescription opioids, and provide referral and treatment, as indicated.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Maternal methadone use in pregnancy: factors associated with the development of neonatal abstinence syndrome and implications for healthcare resources.

            The objectives of this study were to investigate factors associated with the development of neonatal abstinence syndrome (NAS) and to assess the implications for healthcare resources of infants born to drug-misusing women. Retrospective cohort study from 1 January 2004 to 31 December 2006. Inner-city maternity hospital providing dedicated multidisciplinary care to drug-misusing women. Four hundred and fifty singleton pregnancies of drug-misusing women prescribed substitute methadone in pregnancy. Case note review. Development of NAS and duration of infant hospital stay. 45.5% of infants developed NAS requiring pharmacological treatment. The odds ratio of the infant developing NAS was independently related to prescribed maternal methadone dose rather than associated polydrug misuse. Breastfeeding was associated with reduced odds of requiring treatment for NAS (OR 0.55, 95% CI 0.34-0.88). Preterm birth did not influence the odds of the infant receiving treatment for NAS. 48.4% infants were admitted to the neonatal unit (NNU) 40% of these primarily for treatment of NAS. The median total hospital stay for all infants was 10 days (interquartile range 7-17 days). Infants born to methadone-prescribed drug-misusing mothers represented 2.9% of hospital births, but used 18.2% of NNU cot days. Higher maternal methadone dose is associated with a higher incidence of NAS. Pregnant drug-misusing women should be encouraged and supported to breastfeed. Their infants are extremely vulnerable and draw heavily on healthcare resources.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              The Effects of Maternally Administered Methadone, Buprenorphine and Naltrexone on Offspring: Review of Human and Animal Data

              Most women using heroin are of reproductive age with major risks for their infants. We review clinical and experimental data on fetal, neonatal and postnatal complications associated with methadone, the current “gold standard”, and compare these with more recent, but limited, data on developmental effects of buprenorphine, and naltrexone. Methadone is a µ-opioid receptor agonist and is commonly recommended for treatment of opioid dependence during pregnancy. However, it has undesired outcomes including neonatal abstinence syndrome (NAS). Animal studies also indicate detrimental effects on growth, behaviour, neuroanatomy and biochemistry, and increased perinatal mortality. Buprenorphine is a partial µ-opioid receptor agonist and a κ-opioid receptor antagonist. Clinical observations suggest that buprenorphine during pregnancy is similar to methadone on developmental measures but is potentially superior in reducing the incidence and prognosis of NAS. However, small animal studies demonstrate that low doses of buprenorphine during pregnancy and lactation lead to changes in offspring behaviour, neuroanatomy and biochemistry. Naltrexone is a non-selective opioid receptor antagonist. Although data are limited, humans treated with oral or sustained-release implantable naltrexone suggest outcomes potentially superior to those with methadone or buprenorphine. However, animal studies using oral or injectable naltrexone have shown developmental changes following exposure during pregnancy and lactation, raising concerns about its use in humans. Animal studies using chronic exposure, equivalent to clinical depot formulations, are required to evaluate safety. While each treatment is likely to have maternal advantages and disadvantages, studies are urgently required to determine which is optimal for offspring in the short and long term.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                04 May 2023
                2023
                : 14
                : 1182571
                Affiliations
                [1] 1 Auckland Bioengineering Institute , The University of Auckland , Auckland, New Zealand
                [2] 2 Chongqing Food and Drug Control Institute , Chongqing, China
                Author notes

                Edited by: Elena Ramírez, University Hospital La Paz, Spain

                Reviewed by: Tomohiro Nishimura, Keio University, Japan

                Article
                1182571
                10.3389/fphar.2023.1182571
                10192552
                345dc692-a226-49d8-982e-c9fa00395eb6
                Copyright © 2023 Ho and Zhang.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 10 March 2023
                : 18 April 2023
                Funding
                Funded by: Key Technology Innovation Special of Key Industries of the Chongqing Science and Technology Bureau , doi 10.13039/501100019444;
                Award ID: cstc2022jxjl0299
                HH and EZ acknowledge the funding support of Chongqing Science and Technology Bureau (Grant number: cstc2022jxjl0299).
                Categories
                Pharmacology
                Opinion
                Custom metadata
                Translational Pharmacology

                Pharmacology & Pharmaceutical medicine
                methadone,opioids,pbpk,p-glycoprotein,model
                Pharmacology & Pharmaceutical medicine
                methadone, opioids, pbpk, p-glycoprotein, model

                Comments

                Comment on this article