孕期用药:临床药理学外推框架,弥补知识空白。

IF 3.1 3区 医学 Q2 PHARMACOLOGY & PHARMACY
Paola Coppola, Eva Gil Berglund, Karen Rowland Yeo
{"title":"孕期用药:临床药理学外推框架,弥补知识空白。","authors":"Paola Coppola,&nbsp;Eva Gil Berglund,&nbsp;Karen Rowland Yeo","doi":"10.1002/psp4.13242","DOIUrl":null,"url":null,"abstract":"<p>Drug treatment may be required during pregnancy, both for pregnant women and their unborn children. About 6 million pregnancies in the United States (US) occur each year, with most women taking at least one prescription medication during pregnancy and more than half of the mothers taking medicines after delivery (Pregnant?_Breastfeeding?_FDA_Aims_to_Improve_Drug_Information_[fda.gov]). However, in our attempts to protect the unborn children or breastfeeding infants, information to support such treatment is rarely generated and drugs are often used off-label.</p><p>Systematic exclusion of pregnant women from clinical trials at all stages does not allow the collection of data to support the safe use of medicines during pregnancy. Dosing strategies to treat health conditions developed either before or during pregnancy often rely on data from healthy and/or nonpregnant subjects, instead of being driven by complex pregnancy-related physiological changes on drug exposure. Despite the recognized medical need, a recent review captured labels with clinically meaningful interventions in pregnancy for only 139 medications in the US and 20 in the European Union (EU); in both cases, 30%–40% had established doses for pregnant populations.<span><sup>1</sup></span> Information on dosing during pregnancy is often unavailable in original regulatory submissions limiting label recommendation. Pre-authorization data in pregnant population is generally not requested and post-authorization registry studies are mainly required for drugs where substantial use during pregnancy is foreseen, for example, in malaria or for HIV treatment, monitoring pregnancy outcomes in women exposed to drugs during gestation (Postmarket_Requirements_and_Commitments_[fda.gov]). In addition to data collection in registries, when pregnancy is expected to impact systemic drug levels, clinical PK data are generated post-authorization to inform dosing recommendations (e.g., rilpivirine, darunavir, cobicistat).</p><p>For years, the clinical need for drug treatment during pregnancy has been left largely unresolved by regulators and sponsors, leaving the risk–benefit assessment to prescribers and patients. However, health care and community attention to this unmet medical need has resulted in increased regulatory action. In 2018, the FDA published a draft guidance on Scientific and Ethical Considerations for the Inclusion of pregnant women in Clinical Trials. In 2022, current thinking and regulatory efforts were communicated by Sewell et al.<span><sup>2</sup></span> and the FDA diversity plan framework (Diversity_Plans_to_Improve_Enrollment_of_Participants_from_Underrepresented_Racial_and_Ethnic_Populations_in_Clinical_Trials_Guidance_for_Industry_(fda.gov)). Furthermore, regulators from the FDA, EMA, and MHRA acknowledged the urgent need to shift from systematic exclusion to the inclusion of pregnant and breastfeeding women in clinical trials at the International Coalition of Medicines Regulatory Authorities Pregnancy and Lactation Workshop (ICMRA_Pregnancy_and_Lactation_Workshop_International_Coalition_of_Medicines_Regulatory_Authorities_(ICMRA)). It was proposed that applicants should develop and submit a “Maternal Investigation Plan,” outlining the strategy to study these populations. This change of approach requires international collaboration and harmonization, and foundations were laid for the development of the ICH21 Guideline (ICH_E21_Final_Concept_Paper_2023) which will outline the investigational development plan, alongside other factors considered in the extrapolation framework of the draft ICH E11 guideline for pediatrics (draft-ich-guideline-e11a-pediatric-extrapolation-step-2b_en.pdf_(europa.eu)). The FDA has also held workshops to encourage an increase in the number of studies conducted in pregnant woman (Pharmacokinetic_Evaluation_in_Pregnancy_(fda.gov); Fetal_Pharmacology_and_Therapeutics_(fda.gov)).</p><p>Pregnancy-related physiological changes can affect drug PK resulting in possible loss of efficacy, or potential toxicity in both the mother and fetus. Phase I (e.g., CYP3A4, CYP2D6, and CYP219) and phase II (UGT1A1 and UGT1A4) metabolizing enzyme activities are altered during pregnancy. This is illustrated by the observed ~60% increase in clearance and ~60% decrease in exposure of the CYP2D6 substrates metoprolol and fluoxetine, respectively.<span><sup>3, 4</sup></span> Corresponding decreases in exposure have been reported for the CYP3A substrates rilpivirine and cobicistat-boosted darunavir as well as for the UGT1A1 substrate dolutegravir. Increased glomerular filtration rate during pregnancy, can lead to reduced exposure of drugs undergoing renal excretion (e.g., ceftazidime).</p><p>In some cases, the decrease in exposure has been reflected in the labeling, driving either dose recommendations, monitoring, or contraindications.</p><p>In the original rilpivirine label (2011), the use of the drug during pregnancy was restricted to cases where the potential benefit justifies the potential risk. In 2018, the labeling was updated based on new data (i.e., 30%–40% lower exposure in second and third trimesters than postpartum), resulting in a recommendation to closely monitor viral load due to lower exposures during pregnancy (EDURANT®_(rilpivirine)_US_label).</p><p>Similarly, the exposure of dolutegravir was reported to be up to 37% lower during pregnancy.<span><sup>5</sup></span> In the recently updated label (2024), clinical fetal outcome data from observational studies supported the removal of pregnancy testing before initiation of treatment as well as the warning of embryo–fetal toxicity (TIVICAY_(dolutegravir)_US_label).</p><p>As an example of major labeling impact, the exposure of darunavir boosted with cobicistat was substantially lower (&gt;50% decrease in AUC, and ~90% decrease in <i>C</i><sub>min</sub>) during the second and third trimesters. Hence, the label states that use during pregnancy is not recommended. An exposure registry monitors pregnancy outcomes in individuals taking the drugs (PREZCOBIX®_(darunavir_and_cobicistat)_US_label). Publicly available data are not sufficient to allow the evaluation of whether a dose increase may be feasible and safe for the mother and fetus.</p><p>The EMA recommends therapeutic drug monitoring for ritonavir-boosted atazanavir (300/100 mg) as insufficient exposure to atazanavir may occur in pregnant patients. Moreover, both EMA and FDA labels state that if either tenofovir or an H2-receptor antagonist is co-administered in pregnant women, an atazanavir dose increase (400 mg) may be required due to enhanced exposure reduction (REYATAZ_[atazanavir]_US_label; REYATAZ_EMA_SmPC).</p><p>Before these labeling changes were implemented, it is likely that uninformed, off-label use occurred.</p><p>The pediatric extrapolation framework outlined in ICH E11 (ICH_Guideline_E11A_on_Pediatric_Extrapolation) can be adapted to pregnancy, supporting decisions on the need for studies, informing study design and labeling update, and generating new data to fill the knowledge gaps. Particular consideration should be made for teratogenicity and fetal toxicity, and a cautious, informed, approach is recommended. Evaluation and interpretation of fetal and preclinical data informing risk should be considered but it is outside the scope of this paper. Registries provide important follow-up information on fetal, that is, pediatric safety detecting signals and allow broader scientific analysis. If the use of a medicine in pregnant women, during certain trimesters, is too rare for conventional studies to be possible, adapted strategies to obtain PK and clinical information should be planned for. Labeling language could be considered to enable these approaches.</p><p>As in ICH E11, the drug development plan should consider the clinical need for treatment and foreseen risks in each trimester as well as significant benefits compared with available therapies. The efficacy extrapolation approach could be based on similarity in disease and in response to treatment, drug pharmacology, expected similarity in target exposure, and how to reach this exposure in pregnant individuals. Scientific literature, preclinical and clinical data, clinical experience with other drugs, including RWE, and foreseen PK differences, should all be considered (Figure 1).</p><p>The extrapolation framework could support decisions on mother and fetal safety management including study design, sample size (which might be smaller than conventional phase III), generation of post-authorization safety data in a larger population, need for long-term follow-up and registries, etc. Key considerations also include risks to mother and fetus if not treating the disease, the need for gradual recruitment into earlier trimesters, available safety information in nonpregnant women, whether target/off-target effects could be different, or particularly impact pregnant women or the fetus. Safety data available for drugs with similar mechanisms of action, or off-target effects are crucial, as well as preclinical fetal toxicity/teratogenicity information and the need to follow-up specific short- and long-term effects, etc. (Table 1). Expected fetal exposure to drugs should if possible be considered.</p><p>Availability of data assessing similarity of exposure–response (ER) relationships for efficacy and safety compared with nonpregnant should be reflected in the study design, and physiological factors impacting drug exposure during pregnancy considered in dose selection, and significant uncertainties resolved by confirmatory “PK lead-in” investigations.</p><p>Sparse sampling approaches should be optimized using population PK (pop-PK) to support study designs, for example, informing sample size estimations. The use of physiologically-based pharmacokinetic (PBPK) modeling to simulate drug exposure is recommended for initial dose selection, as it allows multifactorial mechanistic knowledge to inform exposure predictions. For marketed drugs, the analysis of pharmacovigilance and real-world data on use during pregnancy should be considered. It is also important to understand whether loss of efficacy is expected due to altered drug systemic exposure, and if temporary dose adjustment is needed.</p><p>The clinical pharmacology strategy for investigating the impact of pregnancy on drug exposure requires integration of available data, including clinical PK data collected in nonpregnant populations, preclinical information, a good understanding of the effect of pregnancy-related physiological changes on drug exposure, potential DDIs, and exposure–response relationship. The totality of evidence can inform the development of a MIDD-based extrapolation framework to identify and fill knowledge gaps underlining a development strategy to optimize the use of available data.</p><p>The use of PBPK models, incorporating relevant physiological changes, to predict drug exposure and PD response (target and off-target pharmacological effects) in pregnant women and the fetus is being considered by regulators as a tool to possibly inform dosing during pregnancy and support benefit–risk decisions [ICH_E21_Final_Concept_Paper_2023].<span><sup>6, 7</sup></span></p><p>When there is a good understanding of the effect of pregnancy on the physiological factors impacting the disposition of the specific drug, PBPK modeling can be confidently applied to predict exposure changes and drive dosing decisions. Several authors showed that PBPK modeling can reliably predict those changes in exposure in pregnancy compared with nonpregnant populations for a range of drugs,<span><sup>8, 9</sup></span> [Pregnancy_Physiologically_Based_Pharmacokinetic_(PBPK)_Modeling_with_Population_Variability_for_Drug_Safety_and_Efficacy_Assessment_(fda.gov)]. A high-fidelity drug model<span><sup>10</sup></span> (where the elimination routes are clearly elucidated) developed initially in the nonpregnant population can then be applied to predict an initial dose in pregnant subjects. Once PK data in the pregnant patients have been accrued, pop-PK modeling could be used to support the PBPK modeling, quantify ER relationships, and analyze covariates affecting exposure.</p><p>A change of mindset appears to have been adopted by the scientific and regulatory community. The need for pregnant women to have well-informed medical treatments has been recognized, going from a protection from research to a protection by research strategy. The risk of fetal toxicity should be reflected in relevant parts of the development and multifactorial risk–benefit discussions. The regulatory framework could be based on approaches like global pediatric regulations. Indeed, we propose to adapt the pediatric development extrapolation framework, (ICH_Guideline_E11A_on_Pediatric_Extrapolation), providing a systematic approach to drug development, identifying important knowledge gaps to be addressed in the clinical study design and follow-up strategies. A clinical pharmacology strategy based on MIDD approaches, such as PBPK and pop-PK is central to this approach.</p><p>No funding was received for this work.</p><p>P.C. is an employee of Certara Italy (Certara Drug Development Solutions). E.B. is an employee of Certara NL (Certara Drug Development Solutions). K.R.Y. is an employee of Certara UK Limited (Simcyp Division).</p><p>As Deputy-Editor-Chief for <i>CPT: Pharmacometrics &amp; Systems Pharmacology</i>, Karen Rowland Yeo was not involved in the review or decision-making processes for this work.</p>","PeriodicalId":10774,"journal":{"name":"CPT: Pharmacometrics & Systems Pharmacology","volume":"13 11","pages":"1830-1834"},"PeriodicalIF":3.1000,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13242","citationCount":"0","resultStr":"{\"title\":\"Medicines in pregnancy: A clinical pharmacology extrapolation framework to address knowledge gaps\",\"authors\":\"Paola Coppola,&nbsp;Eva Gil Berglund,&nbsp;Karen Rowland Yeo\",\"doi\":\"10.1002/psp4.13242\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Drug treatment may be required during pregnancy, both for pregnant women and their unborn children. About 6 million pregnancies in the United States (US) occur each year, with most women taking at least one prescription medication during pregnancy and more than half of the mothers taking medicines after delivery (Pregnant?_Breastfeeding?_FDA_Aims_to_Improve_Drug_Information_[fda.gov]). However, in our attempts to protect the unborn children or breastfeeding infants, information to support such treatment is rarely generated and drugs are often used off-label.</p><p>Systematic exclusion of pregnant women from clinical trials at all stages does not allow the collection of data to support the safe use of medicines during pregnancy. Dosing strategies to treat health conditions developed either before or during pregnancy often rely on data from healthy and/or nonpregnant subjects, instead of being driven by complex pregnancy-related physiological changes on drug exposure. Despite the recognized medical need, a recent review captured labels with clinically meaningful interventions in pregnancy for only 139 medications in the US and 20 in the European Union (EU); in both cases, 30%–40% had established doses for pregnant populations.<span><sup>1</sup></span> Information on dosing during pregnancy is often unavailable in original regulatory submissions limiting label recommendation. Pre-authorization data in pregnant population is generally not requested and post-authorization registry studies are mainly required for drugs where substantial use during pregnancy is foreseen, for example, in malaria or for HIV treatment, monitoring pregnancy outcomes in women exposed to drugs during gestation (Postmarket_Requirements_and_Commitments_[fda.gov]). In addition to data collection in registries, when pregnancy is expected to impact systemic drug levels, clinical PK data are generated post-authorization to inform dosing recommendations (e.g., rilpivirine, darunavir, cobicistat).</p><p>For years, the clinical need for drug treatment during pregnancy has been left largely unresolved by regulators and sponsors, leaving the risk–benefit assessment to prescribers and patients. However, health care and community attention to this unmet medical need has resulted in increased regulatory action. In 2018, the FDA published a draft guidance on Scientific and Ethical Considerations for the Inclusion of pregnant women in Clinical Trials. In 2022, current thinking and regulatory efforts were communicated by Sewell et al.<span><sup>2</sup></span> and the FDA diversity plan framework (Diversity_Plans_to_Improve_Enrollment_of_Participants_from_Underrepresented_Racial_and_Ethnic_Populations_in_Clinical_Trials_Guidance_for_Industry_(fda.gov)). Furthermore, regulators from the FDA, EMA, and MHRA acknowledged the urgent need to shift from systematic exclusion to the inclusion of pregnant and breastfeeding women in clinical trials at the International Coalition of Medicines Regulatory Authorities Pregnancy and Lactation Workshop (ICMRA_Pregnancy_and_Lactation_Workshop_International_Coalition_of_Medicines_Regulatory_Authorities_(ICMRA)). It was proposed that applicants should develop and submit a “Maternal Investigation Plan,” outlining the strategy to study these populations. This change of approach requires international collaboration and harmonization, and foundations were laid for the development of the ICH21 Guideline (ICH_E21_Final_Concept_Paper_2023) which will outline the investigational development plan, alongside other factors considered in the extrapolation framework of the draft ICH E11 guideline for pediatrics (draft-ich-guideline-e11a-pediatric-extrapolation-step-2b_en.pdf_(europa.eu)). The FDA has also held workshops to encourage an increase in the number of studies conducted in pregnant woman (Pharmacokinetic_Evaluation_in_Pregnancy_(fda.gov); Fetal_Pharmacology_and_Therapeutics_(fda.gov)).</p><p>Pregnancy-related physiological changes can affect drug PK resulting in possible loss of efficacy, or potential toxicity in both the mother and fetus. Phase I (e.g., CYP3A4, CYP2D6, and CYP219) and phase II (UGT1A1 and UGT1A4) metabolizing enzyme activities are altered during pregnancy. This is illustrated by the observed ~60% increase in clearance and ~60% decrease in exposure of the CYP2D6 substrates metoprolol and fluoxetine, respectively.<span><sup>3, 4</sup></span> Corresponding decreases in exposure have been reported for the CYP3A substrates rilpivirine and cobicistat-boosted darunavir as well as for the UGT1A1 substrate dolutegravir. Increased glomerular filtration rate during pregnancy, can lead to reduced exposure of drugs undergoing renal excretion (e.g., ceftazidime).</p><p>In some cases, the decrease in exposure has been reflected in the labeling, driving either dose recommendations, monitoring, or contraindications.</p><p>In the original rilpivirine label (2011), the use of the drug during pregnancy was restricted to cases where the potential benefit justifies the potential risk. In 2018, the labeling was updated based on new data (i.e., 30%–40% lower exposure in second and third trimesters than postpartum), resulting in a recommendation to closely monitor viral load due to lower exposures during pregnancy (EDURANT®_(rilpivirine)_US_label).</p><p>Similarly, the exposure of dolutegravir was reported to be up to 37% lower during pregnancy.<span><sup>5</sup></span> In the recently updated label (2024), clinical fetal outcome data from observational studies supported the removal of pregnancy testing before initiation of treatment as well as the warning of embryo–fetal toxicity (TIVICAY_(dolutegravir)_US_label).</p><p>As an example of major labeling impact, the exposure of darunavir boosted with cobicistat was substantially lower (&gt;50% decrease in AUC, and ~90% decrease in <i>C</i><sub>min</sub>) during the second and third trimesters. Hence, the label states that use during pregnancy is not recommended. An exposure registry monitors pregnancy outcomes in individuals taking the drugs (PREZCOBIX®_(darunavir_and_cobicistat)_US_label). Publicly available data are not sufficient to allow the evaluation of whether a dose increase may be feasible and safe for the mother and fetus.</p><p>The EMA recommends therapeutic drug monitoring for ritonavir-boosted atazanavir (300/100 mg) as insufficient exposure to atazanavir may occur in pregnant patients. Moreover, both EMA and FDA labels state that if either tenofovir or an H2-receptor antagonist is co-administered in pregnant women, an atazanavir dose increase (400 mg) may be required due to enhanced exposure reduction (REYATAZ_[atazanavir]_US_label; REYATAZ_EMA_SmPC).</p><p>Before these labeling changes were implemented, it is likely that uninformed, off-label use occurred.</p><p>The pediatric extrapolation framework outlined in ICH E11 (ICH_Guideline_E11A_on_Pediatric_Extrapolation) can be adapted to pregnancy, supporting decisions on the need for studies, informing study design and labeling update, and generating new data to fill the knowledge gaps. Particular consideration should be made for teratogenicity and fetal toxicity, and a cautious, informed, approach is recommended. Evaluation and interpretation of fetal and preclinical data informing risk should be considered but it is outside the scope of this paper. Registries provide important follow-up information on fetal, that is, pediatric safety detecting signals and allow broader scientific analysis. If the use of a medicine in pregnant women, during certain trimesters, is too rare for conventional studies to be possible, adapted strategies to obtain PK and clinical information should be planned for. Labeling language could be considered to enable these approaches.</p><p>As in ICH E11, the drug development plan should consider the clinical need for treatment and foreseen risks in each trimester as well as significant benefits compared with available therapies. The efficacy extrapolation approach could be based on similarity in disease and in response to treatment, drug pharmacology, expected similarity in target exposure, and how to reach this exposure in pregnant individuals. Scientific literature, preclinical and clinical data, clinical experience with other drugs, including RWE, and foreseen PK differences, should all be considered (Figure 1).</p><p>The extrapolation framework could support decisions on mother and fetal safety management including study design, sample size (which might be smaller than conventional phase III), generation of post-authorization safety data in a larger population, need for long-term follow-up and registries, etc. Key considerations also include risks to mother and fetus if not treating the disease, the need for gradual recruitment into earlier trimesters, available safety information in nonpregnant women, whether target/off-target effects could be different, or particularly impact pregnant women or the fetus. Safety data available for drugs with similar mechanisms of action, or off-target effects are crucial, as well as preclinical fetal toxicity/teratogenicity information and the need to follow-up specific short- and long-term effects, etc. (Table 1). Expected fetal exposure to drugs should if possible be considered.</p><p>Availability of data assessing similarity of exposure–response (ER) relationships for efficacy and safety compared with nonpregnant should be reflected in the study design, and physiological factors impacting drug exposure during pregnancy considered in dose selection, and significant uncertainties resolved by confirmatory “PK lead-in” investigations.</p><p>Sparse sampling approaches should be optimized using population PK (pop-PK) to support study designs, for example, informing sample size estimations. The use of physiologically-based pharmacokinetic (PBPK) modeling to simulate drug exposure is recommended for initial dose selection, as it allows multifactorial mechanistic knowledge to inform exposure predictions. For marketed drugs, the analysis of pharmacovigilance and real-world data on use during pregnancy should be considered. It is also important to understand whether loss of efficacy is expected due to altered drug systemic exposure, and if temporary dose adjustment is needed.</p><p>The clinical pharmacology strategy for investigating the impact of pregnancy on drug exposure requires integration of available data, including clinical PK data collected in nonpregnant populations, preclinical information, a good understanding of the effect of pregnancy-related physiological changes on drug exposure, potential DDIs, and exposure–response relationship. The totality of evidence can inform the development of a MIDD-based extrapolation framework to identify and fill knowledge gaps underlining a development strategy to optimize the use of available data.</p><p>The use of PBPK models, incorporating relevant physiological changes, to predict drug exposure and PD response (target and off-target pharmacological effects) in pregnant women and the fetus is being considered by regulators as a tool to possibly inform dosing during pregnancy and support benefit–risk decisions [ICH_E21_Final_Concept_Paper_2023].<span><sup>6, 7</sup></span></p><p>When there is a good understanding of the effect of pregnancy on the physiological factors impacting the disposition of the specific drug, PBPK modeling can be confidently applied to predict exposure changes and drive dosing decisions. Several authors showed that PBPK modeling can reliably predict those changes in exposure in pregnancy compared with nonpregnant populations for a range of drugs,<span><sup>8, 9</sup></span> [Pregnancy_Physiologically_Based_Pharmacokinetic_(PBPK)_Modeling_with_Population_Variability_for_Drug_Safety_and_Efficacy_Assessment_(fda.gov)]. A high-fidelity drug model<span><sup>10</sup></span> (where the elimination routes are clearly elucidated) developed initially in the nonpregnant population can then be applied to predict an initial dose in pregnant subjects. Once PK data in the pregnant patients have been accrued, pop-PK modeling could be used to support the PBPK modeling, quantify ER relationships, and analyze covariates affecting exposure.</p><p>A change of mindset appears to have been adopted by the scientific and regulatory community. The need for pregnant women to have well-informed medical treatments has been recognized, going from a protection from research to a protection by research strategy. The risk of fetal toxicity should be reflected in relevant parts of the development and multifactorial risk–benefit discussions. The regulatory framework could be based on approaches like global pediatric regulations. Indeed, we propose to adapt the pediatric development extrapolation framework, (ICH_Guideline_E11A_on_Pediatric_Extrapolation), providing a systematic approach to drug development, identifying important knowledge gaps to be addressed in the clinical study design and follow-up strategies. A clinical pharmacology strategy based on MIDD approaches, such as PBPK and pop-PK is central to this approach.</p><p>No funding was received for this work.</p><p>P.C. is an employee of Certara Italy (Certara Drug Development Solutions). E.B. is an employee of Certara NL (Certara Drug Development Solutions). K.R.Y. is an employee of Certara UK Limited (Simcyp Division).</p><p>As Deputy-Editor-Chief for <i>CPT: Pharmacometrics &amp; Systems Pharmacology</i>, Karen Rowland Yeo was not involved in the review or decision-making processes for this work.</p>\",\"PeriodicalId\":10774,\"journal\":{\"name\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"volume\":\"13 11\",\"pages\":\"1830-1834\"},\"PeriodicalIF\":3.1000,\"publicationDate\":\"2024-10-14\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/psp4.13242\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"CPT: Pharmacometrics & Systems Pharmacology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/psp4.13242\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"CPT: Pharmacometrics & Systems Pharmacology","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/psp4.13242","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

怀孕期间,孕妇和胎儿都可能需要接受药物治疗。美国每年约有 600 万例妊娠,大多数妇女在怀孕期间至少服用一种处方药,半数以上的母亲在分娩后还在服用药物(Pregnant?_Breastfeeding?_FDA_Aims_to_Improve_Drug_Information_[fda.gov])。然而,为了保护未出生的婴儿或母乳喂养的婴儿,我们很少提供支持这种治疗的信息,药物往往在标签外使用。在临床试验的各个阶段,系统性地将孕妇排除在外,无法收集支持孕期安全用药的数据。在妊娠前或妊娠期间制定的治疗健康状况的剂量策略往往依赖于健康和/或非妊娠受试者的数据,而不是受与妊娠有关的复杂生理变化对药物暴露的影响。尽管存在公认的医疗需求,但最近的一项研究仅发现美国和欧盟分别有 139 种和 20 种药物的标签对妊娠期有临床意义的干预措施;在这两种情况下,30%-40% 的药物为妊娠人群确定了剂量1 。一般不要求提供妊娠人群的授权前数据,授权后登记研究主要针对预计在妊娠期间大量使用的药物,例如疟疾或艾滋病治疗药物,监测妊娠期间接触药物的妇女的妊娠结局(Postmarket_Requirements_and_Commitments_[fda.gov])。除了在登记册中收集数据外,当预计妊娠会影响全身用药水平时,还会在获得授权后生成临床 PK 数据,以便为用药建议提供依据(如利匹韦林、达鲁那韦、考比司他)。多年来,监管机构和申办者在很大程度上一直没有解决妊娠期药物治疗的临床需求问题,而是将风险效益评估留给了处方者和患者。然而,医疗保健和社会各界对这一尚未满足的医疗需求的关注已导致监管行动的增加。2018 年,FDA 发布了《将孕妇纳入临床试验的科学和伦理考虑因素》指南草案。2022 年,Sewell 等人2 和 FDA 多样性计划框架(Diversity_Plans_to_Improve_Enrollment_of_Participants_from_Underrepresented_Racial_and_Ethnic_Populations_in_Clinical_Trials_Guidance_for_Industry_(fda.gov))交流了当前的思路和监管工作。此外,在国际药品监管机构联盟妊娠和哺乳期研讨会(ICMRA_Pregnancy_and_Lactation_Workshop_International_Coalition_of_Medicines_Regulatory_Authorities_(ICMRA))上,来自 FDA、EMA 和 MHRA 的监管者承认,迫切需要将系统性排斥转变为将孕妇和哺乳期妇女纳入临床试验。会议建议,申请者应制定并提交一份 "孕产妇调查计划",概述研究这些人群的策略。这种方法的改变需要国际合作与协调,并为 ICH21 指导原则(ICH_E21_Final_Concept_Paper_2023)的制定奠定了基础,该指导原则将概述研究开发计划,以及 ICH E11 指导原则草案儿科外推框架中考虑的其他因素(draft-ich-guideline-e11a-pediatric-extrapolation-step-2b_en.pdf_(europa.eu))。与妊娠有关的生理变化会影响药物的 PK 值,从而可能导致药效丧失,或对母亲和胎儿产生潜在毒性。妊娠期的 I 期(如 CYP3A4、CYP2D6 和 CYP219)和 II 期(UGT1A1 和 UGT1A4)代谢酶活性会发生变化。CYP2D6底物美托洛尔(metoprolol)和氟西汀(fluoxetine)的清除率和暴露量分别增加了约 60%和减少了约 60%,3, 4 CYP3A 底物利匹韦林(rilpivirine)和科比司他(cobicistat-boosted darunavir)以及 UGT1A1 底物多鲁曲韦 (dolutegravir)的暴露量也相应减少。妊娠期间肾小球滤过率的增加可导致经肾脏排泄的药物(如头孢唑肟)的暴露量减少。在某些情况下,暴露量的减少已反映在标签中,推动了剂量建议、监测或禁忌。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

Medicines in pregnancy: A clinical pharmacology extrapolation framework to address knowledge gaps

Medicines in pregnancy: A clinical pharmacology extrapolation framework to address knowledge gaps

Drug treatment may be required during pregnancy, both for pregnant women and their unborn children. About 6 million pregnancies in the United States (US) occur each year, with most women taking at least one prescription medication during pregnancy and more than half of the mothers taking medicines after delivery (Pregnant?_Breastfeeding?_FDA_Aims_to_Improve_Drug_Information_[fda.gov]). However, in our attempts to protect the unborn children or breastfeeding infants, information to support such treatment is rarely generated and drugs are often used off-label.

Systematic exclusion of pregnant women from clinical trials at all stages does not allow the collection of data to support the safe use of medicines during pregnancy. Dosing strategies to treat health conditions developed either before or during pregnancy often rely on data from healthy and/or nonpregnant subjects, instead of being driven by complex pregnancy-related physiological changes on drug exposure. Despite the recognized medical need, a recent review captured labels with clinically meaningful interventions in pregnancy for only 139 medications in the US and 20 in the European Union (EU); in both cases, 30%–40% had established doses for pregnant populations.1 Information on dosing during pregnancy is often unavailable in original regulatory submissions limiting label recommendation. Pre-authorization data in pregnant population is generally not requested and post-authorization registry studies are mainly required for drugs where substantial use during pregnancy is foreseen, for example, in malaria or for HIV treatment, monitoring pregnancy outcomes in women exposed to drugs during gestation (Postmarket_Requirements_and_Commitments_[fda.gov]). In addition to data collection in registries, when pregnancy is expected to impact systemic drug levels, clinical PK data are generated post-authorization to inform dosing recommendations (e.g., rilpivirine, darunavir, cobicistat).

For years, the clinical need for drug treatment during pregnancy has been left largely unresolved by regulators and sponsors, leaving the risk–benefit assessment to prescribers and patients. However, health care and community attention to this unmet medical need has resulted in increased regulatory action. In 2018, the FDA published a draft guidance on Scientific and Ethical Considerations for the Inclusion of pregnant women in Clinical Trials. In 2022, current thinking and regulatory efforts were communicated by Sewell et al.2 and the FDA diversity plan framework (Diversity_Plans_to_Improve_Enrollment_of_Participants_from_Underrepresented_Racial_and_Ethnic_Populations_in_Clinical_Trials_Guidance_for_Industry_(fda.gov)). Furthermore, regulators from the FDA, EMA, and MHRA acknowledged the urgent need to shift from systematic exclusion to the inclusion of pregnant and breastfeeding women in clinical trials at the International Coalition of Medicines Regulatory Authorities Pregnancy and Lactation Workshop (ICMRA_Pregnancy_and_Lactation_Workshop_International_Coalition_of_Medicines_Regulatory_Authorities_(ICMRA)). It was proposed that applicants should develop and submit a “Maternal Investigation Plan,” outlining the strategy to study these populations. This change of approach requires international collaboration and harmonization, and foundations were laid for the development of the ICH21 Guideline (ICH_E21_Final_Concept_Paper_2023) which will outline the investigational development plan, alongside other factors considered in the extrapolation framework of the draft ICH E11 guideline for pediatrics (draft-ich-guideline-e11a-pediatric-extrapolation-step-2b_en.pdf_(europa.eu)). The FDA has also held workshops to encourage an increase in the number of studies conducted in pregnant woman (Pharmacokinetic_Evaluation_in_Pregnancy_(fda.gov); Fetal_Pharmacology_and_Therapeutics_(fda.gov)).

Pregnancy-related physiological changes can affect drug PK resulting in possible loss of efficacy, or potential toxicity in both the mother and fetus. Phase I (e.g., CYP3A4, CYP2D6, and CYP219) and phase II (UGT1A1 and UGT1A4) metabolizing enzyme activities are altered during pregnancy. This is illustrated by the observed ~60% increase in clearance and ~60% decrease in exposure of the CYP2D6 substrates metoprolol and fluoxetine, respectively.3, 4 Corresponding decreases in exposure have been reported for the CYP3A substrates rilpivirine and cobicistat-boosted darunavir as well as for the UGT1A1 substrate dolutegravir. Increased glomerular filtration rate during pregnancy, can lead to reduced exposure of drugs undergoing renal excretion (e.g., ceftazidime).

In some cases, the decrease in exposure has been reflected in the labeling, driving either dose recommendations, monitoring, or contraindications.

In the original rilpivirine label (2011), the use of the drug during pregnancy was restricted to cases where the potential benefit justifies the potential risk. In 2018, the labeling was updated based on new data (i.e., 30%–40% lower exposure in second and third trimesters than postpartum), resulting in a recommendation to closely monitor viral load due to lower exposures during pregnancy (EDURANT®_(rilpivirine)_US_label).

Similarly, the exposure of dolutegravir was reported to be up to 37% lower during pregnancy.5 In the recently updated label (2024), clinical fetal outcome data from observational studies supported the removal of pregnancy testing before initiation of treatment as well as the warning of embryo–fetal toxicity (TIVICAY_(dolutegravir)_US_label).

As an example of major labeling impact, the exposure of darunavir boosted with cobicistat was substantially lower (>50% decrease in AUC, and ~90% decrease in Cmin) during the second and third trimesters. Hence, the label states that use during pregnancy is not recommended. An exposure registry monitors pregnancy outcomes in individuals taking the drugs (PREZCOBIX®_(darunavir_and_cobicistat)_US_label). Publicly available data are not sufficient to allow the evaluation of whether a dose increase may be feasible and safe for the mother and fetus.

The EMA recommends therapeutic drug monitoring for ritonavir-boosted atazanavir (300/100 mg) as insufficient exposure to atazanavir may occur in pregnant patients. Moreover, both EMA and FDA labels state that if either tenofovir or an H2-receptor antagonist is co-administered in pregnant women, an atazanavir dose increase (400 mg) may be required due to enhanced exposure reduction (REYATAZ_[atazanavir]_US_label; REYATAZ_EMA_SmPC).

Before these labeling changes were implemented, it is likely that uninformed, off-label use occurred.

The pediatric extrapolation framework outlined in ICH E11 (ICH_Guideline_E11A_on_Pediatric_Extrapolation) can be adapted to pregnancy, supporting decisions on the need for studies, informing study design and labeling update, and generating new data to fill the knowledge gaps. Particular consideration should be made for teratogenicity and fetal toxicity, and a cautious, informed, approach is recommended. Evaluation and interpretation of fetal and preclinical data informing risk should be considered but it is outside the scope of this paper. Registries provide important follow-up information on fetal, that is, pediatric safety detecting signals and allow broader scientific analysis. If the use of a medicine in pregnant women, during certain trimesters, is too rare for conventional studies to be possible, adapted strategies to obtain PK and clinical information should be planned for. Labeling language could be considered to enable these approaches.

As in ICH E11, the drug development plan should consider the clinical need for treatment and foreseen risks in each trimester as well as significant benefits compared with available therapies. The efficacy extrapolation approach could be based on similarity in disease and in response to treatment, drug pharmacology, expected similarity in target exposure, and how to reach this exposure in pregnant individuals. Scientific literature, preclinical and clinical data, clinical experience with other drugs, including RWE, and foreseen PK differences, should all be considered (Figure 1).

The extrapolation framework could support decisions on mother and fetal safety management including study design, sample size (which might be smaller than conventional phase III), generation of post-authorization safety data in a larger population, need for long-term follow-up and registries, etc. Key considerations also include risks to mother and fetus if not treating the disease, the need for gradual recruitment into earlier trimesters, available safety information in nonpregnant women, whether target/off-target effects could be different, or particularly impact pregnant women or the fetus. Safety data available for drugs with similar mechanisms of action, or off-target effects are crucial, as well as preclinical fetal toxicity/teratogenicity information and the need to follow-up specific short- and long-term effects, etc. (Table 1). Expected fetal exposure to drugs should if possible be considered.

Availability of data assessing similarity of exposure–response (ER) relationships for efficacy and safety compared with nonpregnant should be reflected in the study design, and physiological factors impacting drug exposure during pregnancy considered in dose selection, and significant uncertainties resolved by confirmatory “PK lead-in” investigations.

Sparse sampling approaches should be optimized using population PK (pop-PK) to support study designs, for example, informing sample size estimations. The use of physiologically-based pharmacokinetic (PBPK) modeling to simulate drug exposure is recommended for initial dose selection, as it allows multifactorial mechanistic knowledge to inform exposure predictions. For marketed drugs, the analysis of pharmacovigilance and real-world data on use during pregnancy should be considered. It is also important to understand whether loss of efficacy is expected due to altered drug systemic exposure, and if temporary dose adjustment is needed.

The clinical pharmacology strategy for investigating the impact of pregnancy on drug exposure requires integration of available data, including clinical PK data collected in nonpregnant populations, preclinical information, a good understanding of the effect of pregnancy-related physiological changes on drug exposure, potential DDIs, and exposure–response relationship. The totality of evidence can inform the development of a MIDD-based extrapolation framework to identify and fill knowledge gaps underlining a development strategy to optimize the use of available data.

The use of PBPK models, incorporating relevant physiological changes, to predict drug exposure and PD response (target and off-target pharmacological effects) in pregnant women and the fetus is being considered by regulators as a tool to possibly inform dosing during pregnancy and support benefit–risk decisions [ICH_E21_Final_Concept_Paper_2023].6, 7

When there is a good understanding of the effect of pregnancy on the physiological factors impacting the disposition of the specific drug, PBPK modeling can be confidently applied to predict exposure changes and drive dosing decisions. Several authors showed that PBPK modeling can reliably predict those changes in exposure in pregnancy compared with nonpregnant populations for a range of drugs,8, 9 [Pregnancy_Physiologically_Based_Pharmacokinetic_(PBPK)_Modeling_with_Population_Variability_for_Drug_Safety_and_Efficacy_Assessment_(fda.gov)]. A high-fidelity drug model10 (where the elimination routes are clearly elucidated) developed initially in the nonpregnant population can then be applied to predict an initial dose in pregnant subjects. Once PK data in the pregnant patients have been accrued, pop-PK modeling could be used to support the PBPK modeling, quantify ER relationships, and analyze covariates affecting exposure.

A change of mindset appears to have been adopted by the scientific and regulatory community. The need for pregnant women to have well-informed medical treatments has been recognized, going from a protection from research to a protection by research strategy. The risk of fetal toxicity should be reflected in relevant parts of the development and multifactorial risk–benefit discussions. The regulatory framework could be based on approaches like global pediatric regulations. Indeed, we propose to adapt the pediatric development extrapolation framework, (ICH_Guideline_E11A_on_Pediatric_Extrapolation), providing a systematic approach to drug development, identifying important knowledge gaps to be addressed in the clinical study design and follow-up strategies. A clinical pharmacology strategy based on MIDD approaches, such as PBPK and pop-PK is central to this approach.

No funding was received for this work.

P.C. is an employee of Certara Italy (Certara Drug Development Solutions). E.B. is an employee of Certara NL (Certara Drug Development Solutions). K.R.Y. is an employee of Certara UK Limited (Simcyp Division).

As Deputy-Editor-Chief for CPT: Pharmacometrics & Systems Pharmacology, Karen Rowland Yeo was not involved in the review or decision-making processes for this work.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
5.00
自引率
11.40%
发文量
146
审稿时长
8 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信