151
|
Griffin BL, Stone RH, El-Ibiary SY, Westberg S, Shealy K, Forinash A, Yancey A, Vest K, Karaoui LR, Rafie S, Horlen C, Lodise N, Cieri-Hutcherson N, McBane S, Simonyan A. Guide for Drug Selection During Pregnancy and Lactation: What Pharmacists Need to Know for Current Practice. Ann Pharmacother 2018. [PMID: 29519141 DOI: 10.1177/1060028018764447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To provide guidance for clinicians on risk assessment of medication use during pregnancy and lactation. DATA SOURCES Authors completed PubMed searches to identify articles focused on the use of medications in pregnancy, including fetal development, drug transfer across the placenta, trimester exposure, chronic conditions in pregnancy, medications in lactation, and lactation and chronic disease. STUDY SELECTION AND DATA EXTRACTION Articles were reviewed to provide overall guidance to medication selection during pregnancy. The following information was reviewed: medication use in pregnancy, including fetal development, drug transfer across the placenta, trimester exposure, chronic conditions in pregnancy, medications in lactation, and lactation and chronic disease. DATA SYNTHESIS This article will provide an overview of medication safety considerations during pregnancy and lactation. Information was interpreted to help clinicians predict the potential risk and benefit in each patient to make an evidence-based decision. The article concludes with guidance on risk assessment and how pharmacists may support fellow health care providers and their patients when considering medication use. CONCLUSIONS Information about the effects of medication use during reproductive periods is limited. With the removal of the Food and Drug Administration pregnancy categories, clinicians will be relying on pharmacists to aid in the appropriate selection of therapies for patients. It is critical that pharmacists keep abreast of resources available and be able to assess data to help prescribers and their patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sally Rafie
- 8 University of California San Diego, San Diego, CA, USA
| | - Cheryl Horlen
- 9 University of the Incarnate Word, San Antonio, TX, USA
| | - Nicole Lodise
- 10 Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| | | | | | | |
Collapse
|
152
|
Liao MZ, Gao C, Phillips BR, Neradugomma NK, Han LW, Bhatt DK, Prasad B, Shen DD, Mao Q. Pregnancy Increases Norbuprenorphine Clearance in Mice by Induction of Hepatic Glucuronidation. Drug Metab Dispos 2018; 46:100-108. [PMID: 29158248 PMCID: PMC5765905 DOI: 10.1124/dmd.117.076745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/17/2017] [Indexed: 01/09/2023] Open
Abstract
Norbuprenorphine (NBUP) is the major active metabolite of buprenorphine (BUP) that is commonly used to treat opiate addiction during pregnancy; it possesses 25% of BUP's analgesic activity and 10 times BUP's respiratory depression effect. To optimize BUP's dosing regimen during pregnancy with better efficacy and safety, it is important to understand how pregnancy affects NBUP disposition. In this study, we examined the pharmacokinetics of NBUP in pregnant and nonpregnant mice by administering the same amount of NBUP through retro-orbital injection. We demonstrated that the systemic clearance (CL) of NBUP in pregnant mice increased ∼2.5-fold compared with nonpregnant mice. Intrinsic CL of NBUP by glucuronidation in mouse liver microsomes from pregnant mice was ∼2 times greater than that from nonpregnant mice. Targeted liquid chromatography tandem-mass spectrometry proteomics quantification revealed that hepatic Ugt1a1 and Ugt2b1 protein levels in the same amount of total liver membrane proteins were significantly increased by ∼50% in pregnant mice versus nonpregnant mice. After scaling to the whole liver with consideration of the increase in liver protein content and liver weight, we found that the amounts of Ugt1a1, Ugt1a10, Ugt2b1, and Ugt2b35 protein in the whole liver of pregnant mice were significantly increased ∼2-fold compared with nonpregnant mice. These data suggest that the increased systemic CL of NBUP in pregnant mice is likely caused by an induction of hepatic Ugt expression and activity. The data provide a basis for further mechanistic analysis of pregnancy-induced changes in the disposition of NBUP and drugs that are predominately and extensively metabolized by Ugts.
Collapse
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Brian R Phillips
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Naveen K Neradugomma
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Lyrialle W Han
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Deepak Kumar Bhatt
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
153
|
Pharmacokinetics, Antiviral Activity, and Safety of Rilpivirine in Pregnant Women with HIV-1 Infection: Results of a Phase 3b, Multicenter, Open-Label Study. Infect Dis Ther 2018; 7:147-159. [PMID: 29335895 PMCID: PMC5840103 DOI: 10.1007/s40121-017-0184-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Indexed: 11/01/2022] Open
Abstract
INTRODUCTION Physiologic changes during pregnancy may impact the pharmacokinetics of drugs. In addition, efficacy and safety/tolerability concerns have been identified for some antiretroviral agents. METHODS Human immunodeficiency virus (HIV)-1-infected pregnant women (18-26 weeks gestation) receiving the non-nucleoside reverse transcriptase inhibitor rilpivirine 25 mg once daily were enrolled in this phase 3b, open-label study examining the impact of pregnancy on the pharmacokinetics of rilpivirine when it is given in combination with other antiretroviral agents. Blood samples (collected over the 24-h dosing interval) to assess total and unbound rilpivirine plasma concentrations were obtained during the second and third trimesters (24-28 and 34-38 weeks gestation, respectively) and 6-12 weeks postpartum. Pharmacokinetic parameters were derived using noncompartmental analysis and compared (pregnancy versus postpartum) using linear mixed effects modeling. Antiviral and immunologic response and safety were assessed. RESULTS Nineteen women were enrolled; 15 had evaluable pharmacokinetic results. Total rilpivirine exposure was 29-31% lower during pregnancy versus postpartum; differences were less pronounced for unbound (pharmacodynamically active) rilpivirine. At study entry, 12/19 (63.2%) women were virologically suppressed; 10/12 (83.3%) women were suppressed at the postpartum visit. Twelve infants were born to the 12 women who completed the study (7 discontinued); no perinatal viral transmission was observed among 10 infants with available data. Rilpivirine was generally safe and well tolerated in women and infants exposed in utero. CONCLUSION Despite decreased rilpivirine exposure during pregnancy, treatment was effective in preventing mother-to-child transmission and suppressing HIV-1 RNA in pregnant women. Results suggest that rilpivirine 25 mg once daily, as part of individualized combination antiretroviral therapy, may be an appropriate option for HIV-1-infected pregnant women. TRIAL REGISTRATION ClinicalTrials.gov Identifier, NCT00855335.
Collapse
|
154
|
Prevalence and determinants of vitamin D deficiency in the third trimester of pregnancy: a multicentre study in Switzerland. Br J Nutr 2018; 119:299-309. [PMID: 29318983 DOI: 10.1017/s0007114517003634] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vitamin D deficiency during pregnancy is associated with negative health consequences for mothers and their infants. Data on the vitamin D status of pregnant women in Switzerland are scarce. A three-centre study was conducted in the obstetric departments of Zurich, Bellinzona and Samedan (Switzerland) to investigate the prevalence and determinants of vitamin D deficiency (serum 25-hydroxyvitamin D (25(OH)D)<50 nmol/l) in 3rd-trimester pregnant women living in Switzerland (n 305), and the correlation between 25(OH)D in pregnant women and their offspring at birth (n 278). Demographic and questionnaire data were used to explore the determinants of vitamin D deficiency. Median concentration of serum 25(OH)D in the third trimester of pregnancy was 46·0 nmol/l (1st-3rd quartiles: 30·5-68·5), representing a 53·4 % prevalence of vitamin D deficiency. 25(OH)D levels in the umbilcal cord blood (median: 50·0 nmol/l; 1st-3rd quartiles: 31·0-76·6) strongly correlated with mothers' serum 25(OH)D (Spearman's correlation ρ=0·79, P<0·001). Multivariable logistic regression analysis showed that significant determinants of vitamin D deficiency in pregnant women were centre of study, country of origin, season of delivery and vitamin D supplement intake. Near-term BMI, skin colour, use of sunscreen and mothers' education, although each not individually significant, collectively improved the ability of the model to explain vitamin D status. Low vitamin D levels were common in this sample of pregnant women and their newborns' cord blood. Vitamin D supplement intake was the most actionable determinant of vitamin D status, suggesting that vitamin D supplementation during pregnancy should receive more attention in clinical practice.
Collapse
|
155
|
Wu L, Hafiz MZ, Guan Y, He S, Xiong J, Liu W, Yan B, Li X, Yang J. 17β-estradiol suppresses carboxylesterases by activating c-Jun/AP-1 pathway in primary human and mouse hepatocytes. Eur J Pharmacol 2018; 819:98-107. [DOI: 10.1016/j.ejphar.2017.11.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/16/2017] [Accepted: 11/21/2017] [Indexed: 11/16/2022]
|
156
|
El-Shaarawy AM, Asfour MS, Rashwan DA, Amer MM, El-Menshawe SF, Elkomy MH. Comparison of Three Different Concentrations of Levobupivacaine for Epidural Labor Analgesia: Clinical Effect and Pharmacokinetic Profile. Anesth Essays Res 2018; 12:60-66. [PMID: 29628556 PMCID: PMC5872895 DOI: 10.4103/aer.aer_145_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background: The aim is to compare the clinical effect of three different concentrations of levobupivacaine (0.25%, 0.125%, and 0.0625%) on the sensory and motor block characteristics and mode of delivery during epidural labor analgesia. We also studied the pharmacokinetic profile of the three concentrations during labor. Materials and Methods: Sixty pregnant females undergoing normal vaginal delivery under epidural analgesia were divided into three groups according to the concentration of levobupivacaine used. All parturients received an epidural bolus dose of 15 ml of the desired concentration followed by a continuous infusion of the same concentration at 10 mL/h, each combined with fentanyl 2 μg/mL. Sensory block was assessed by the visual analog score (VAS), whereas motor block was evaluated by the Bromage score. Assessments were performed every 5 min in the first 20 min after initiation of epidural analgesia and then at 30 min interval. The incidence of instrumental delivery and cesarean section was also recorded. The total plasma concentrations of levobupivacaine were determined before the start of epidural analgesia, 5 and 10 min after starting the infusion, at infusion stop time, and 3–8 h after infusion termination. Results: The VAS was significantly lower with levobupivacaine concentrations of 0.25% and 0.125% than 0.0625%. Motor block in the form of Bromage score 1 was observed in 39% of parturients receiving levobupivacaine 0.25% of which 43% were converted to cesarean delivery. No motor block was observed with the other two concentrations. Levobupivacaine peak plasma concentrations increased with increasing the concentration of the local anesthetic. There was no difference in other pharmacokinetic parameters between the three groups. Conclusion: levobupivacaine concentration of 0.125% is superior to other concentrations for epidural labor analgesia as it provides adequate analgesia without motor affection which reflects in a lower incidence of instrumental delivery or cesarean section.
Collapse
Affiliation(s)
| | - Mohamed S Asfour
- Department of Anesthesia, Faculty of Medicine, Beni Suef University, Beni Suef, Egypt
| | - Doaa A Rashwan
- Department of Anesthesia, Faculty of Medicine, Beni Suef University, Beni Suef, Egypt
| | - Mahmoud M Amer
- Department of Anesthesia, Faculty of Medicine, Beni Suef University, Beni Suef, Egypt
| | - Shahira F El-Menshawe
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt
| | - Mohammed H Elkomy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt
| |
Collapse
|
157
|
Quinney SK, Gullapelli R, Haas DM. Translational Systems Pharmacology Studies in Pregnant Women. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 7:69-81. [PMID: 29239132 PMCID: PMC5824114 DOI: 10.1002/psp4.12269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/06/2017] [Accepted: 11/07/2017] [Indexed: 12/26/2022]
Abstract
Pregnancy involves rapid physiological adaptation and complex interplay between mother and fetus. New analytic technologies provide large amounts of genomic, proteomic, and metabolomics data. The integration of these data through bioinformatics, statistical, and systems pharmacology techniques can improve our understanding of the mechanisms of normal maternal physiologic changes and fetal development. New insights into the mechanisms of pregnancy‐related disorders, such as preterm birth (PTB), may lead to the development of new therapeutic interventions and novel biomarkers.
Collapse
Affiliation(s)
- Sara K Quinney
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rakesh Gullapelli
- School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - David M Haas
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
158
|
Schalkwijk S, Greupink R, Burger D. Free dug concentrations in pregnancy: Bound to measure unbound? Br J Clin Pharmacol 2017; 83:2595-2598. [PMID: 28983934 DOI: 10.1111/bcp.13432] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/11/2017] [Accepted: 08/26/2017] [Indexed: 01/17/2023] Open
Affiliation(s)
- Stein Schalkwijk
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pharmacology & Toxicology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rick Greupink
- Department of Pharmacology & Toxicology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - David Burger
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
159
|
Westin AA, Brekke M, Molden E, Skogvoll E, Castberg I, Spigset O. Treatment With Antipsychotics in Pregnancy: Changes in Drug Disposition. Clin Pharmacol Ther 2017. [PMID: 28643331 PMCID: PMC5836849 DOI: 10.1002/cpt.770] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although pregnancy is known to cause changes in drug pharmacokinetics, little is known about its impact on serum levels of antipsychotics. In this study we retrospectively assessed 201 routine serum antipsychotic therapeutic drug monitoring concentration measurements obtained from a total of 110 pregnancies in 103 women, and 512 measurements from the same women before and after pregnancy. Serum concentrations in the third trimester were significantly lower than baseline for quetiapine (-76%; confidence interval (CI), -83%, -66%; P < 0.001) and aripiprazole (-52%; CI, -62%, -39%; P < 0.001), but not for olanzapine (-9%; CI, -28%, +14%; P = 0.40). For the remaining antipsychotics (perphenazine, haloperidol, ziprasidone, risperidone, and clozapine), our dataset was limited, but it indicates that concentrations may decline at least for perphenazine and possibly also for haloperidol. Even though the clinical consequence of the serum concentrations decline remains to be elucidated, our results warrant close clinical monitoring throughout pregnancy, preferentially supported by therapeutic drug monitoring.
Collapse
Affiliation(s)
- Andreas A Westin
- Department of Clinical Pharmacology, St Olav University Hospital, Trondheim, Norway
| | - Malin Brekke
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway.,Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Norway
| | - Eirik Skogvoll
- Department of Anaesthesiology and Intensive Care, St. Olav University Hospital, Trondheim, Norway.,Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingrid Castberg
- Department of Psychiatry, St Olav University Hospital, Trondheim, Norway
| | - Olav Spigset
- Department of Clinical Pharmacology, St Olav University Hospital, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
160
|
Marsousi N, Desmeules JA, Rudaz S, Daali Y. Usefulness of PBPK Modeling in Incorporation of Clinical Conditions in Personalized Medicine. J Pharm Sci 2017; 106:2380-2391. [DOI: 10.1016/j.xphs.2017.04.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/14/2022]
|
161
|
Gómez-Martínez LE. Gestational Age Dependency in the Prenatal Toxicity and in the Disposition Kinetics of the Novel Anticonvulsant HEPP (D,L-3-Hydroxy-3-ethyl-3-phenylpropionamide) after Subcutaneous Administration in Pregnant Rats. Int J Toxicol 2017; 26:237-46. [PMID: 17564905 DOI: 10.1080/10915810701352846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
HEPP (D,L-3-hydroxy-3-ethyl-3-phenylpropionamide) is a novel anticonvulsant with promising anticonvulsant profile, which is being actively researched. The potential maternal and embryo/fetal toxicities of HEPP were evaluated in pregnant rats following subcutaneous (s.c.) administration during organogenesis (gestation days 6 through 14, GDs 6–14) and the fetal period (GDs 14–21). Single- and multiple-dose pharmacokinetics were also evaluated at the same periods in order to establish possible correlations with some maternal or embryo/fetal toxicity end points. Embryotoxicity was mainly indicated by a significant dose-concentration dependency in the increase in resorptions, high percentage of fully resorbed litters, and decrease in embryo body weights during the GD6–14 dosing period. No gross external alterations were observed in live fetuses. There was no indication of maternal toxicity; but a marked increase in maternal body weight was evident following dosing from GD14 to GD21. The maternal plasma profile following single subcutaneous dose of 50 mg/kg on both GD14 and GD21 showed a monoexponential elimination pattern. Statistically significant differences between treatments (GD14 versus GD21) were observed in elimination ( kel = 0.12 versus 0.15 h−1), absorption ( ka = 2.01 versus 3.14 h−1), maximum plasma concentration time points ( Tmax = 1.49 versus 1.01 h); maximum plasma concentration ( Cmax = 40.23 versus 36.31 μg/ml) and areas under the concentration-time curve (AUCs0– ∞ = 421.88 versus 274 μg h/ml. Based on comparisons of Cmax, Tmax, and AUCs0– ∞ between the actual data and single intraperitoneal (i.p.) data previously published, the s.c. administration exhibited slower disposition and higher absorbed amount. After multiple-dose administrations of 50 and 100 mg/kg every 12 h (07:00 and 19:00 h), steady-state plasma levels were lower than the computer prediction, and only slight accumulation was observed. In both dosing periods HEPP levels were similar in mothers and offspring at steady-state conditions. The high incidence of embryo death and reduced embryo weight at GD6–14 dosing compared to GD14–21 dosing suggest that embryos are more sensitive to the deleterious effects of HEPP than fetuses; however, the faster elimination observed at late gestation could also contribute to the lower toxicity observed during the fetal period. Because the maternal HEPP plasma levels and the AUC values were positively correlated with embryo/fetal toxicity end points, both pharmacokinetic parameters could be reliable indicators of offspring exposure and consequently of potential toxicity. These data suggest that the length of time that HEPP is present in the maternal plasma at a sufficiently high concentration could be determinant of adverse effects in the offspring.
Collapse
Affiliation(s)
- Lisbeth E Gómez-Martínez
- Departamento de Recursos del Mar, Unidad Mérida, Centro de Investigación y Estudios Avanzados-CINVESTAV, Instituto Politécnico Nacional, Mérida, Yucatán, México.
| |
Collapse
|
162
|
Zhang Z, Unadkat JD. Development of a Novel Maternal-Fetal Physiologically Based Pharmacokinetic Model II: Verification of the model for passive placental permeability drugs. Drug Metab Dispos 2017; 45:939-946. [PMID: 28049636 PMCID: PMC5506455 DOI: 10.1124/dmd.116.073957] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 12/29/2016] [Indexed: 01/01/2023] Open
Abstract
Fetal exposure to drugs cannot be readily estimated from single time point cord blood sampling at the time of delivery. Therefore, we developed a physiologically based pharmacokinetic (PBPK) model to estimate fetal drug exposure throughout pregnancy. In this study, we report verification of this novel maternal-fetal PBPK (m-f-PBPK) model for drugs that passively diffuse across the placenta and are not metabolized/transported there. Our recently built m-f-PBPK model was populated with gestational age-dependent changes in maternal drug disposition and maternal-fetal physiology. Using midazolam as an in vivo calibrator, the transplacental passive diffusion clearance of theophylline and zidovudine was first estimated. Then, for verification, the predicted maternal plasma (MP) and umbilical venous (UV) plasma drug concentrations by our m-f-PBPK were compared against those observed at term. Overall, our m-f-PBPK model well predicted the maternal and fetal exposure to the two verification drugs, theophylline and zidovudine, at term, across a range of dosing regimens, with nearly all observed MP and UV plasma drug concentrations falling within the 90% prediction interval [i.e., 5th-95th percentile range of a virtual pregnant population (n = 100)]. Prediction precision and bias of theophylline MP and UV were 14.5% and 12.4%, and 9.4% and 7.5%, respectively. Furthermore, for zidovudine, after the exclusion of one unexpectedly low MP concentration, prediction precision and bias for MP and UV were 50.3% and 30.2, and 28.3% and 15.0%, respectively. This m-f-PBPK should be useful to predict fetal exposure to drugs, throughout pregnancy, for drugs that passively diffuse across the placenta.
Collapse
Affiliation(s)
- Zufei Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
163
|
Zhang Z, Imperial MZ, Patilea-Vrana GI, Wedagedera J, Gaohua L, Unadkat JD. Development of a Novel Maternal-Fetal Physiologically Based Pharmacokinetic Model I: Insights into Factors that Determine Fetal Drug Exposure through Simulations and Sensitivity Analyses. Drug Metab Dispos 2017; 45:920-938. [PMID: 28588050 PMCID: PMC5506457 DOI: 10.1124/dmd.117.075192] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 05/25/2017] [Indexed: 12/21/2022] Open
Abstract
Determining fetal drug exposure (except at the time of birth) is not possible for both logistical and ethical reasons. Therefore, we developed a novel maternal-fetal physiologically based pharmacokinetic (m-f-PBPK) model to predict fetal exposure to drugs and populated this model with gestational age-dependent changes in maternal-fetal physiology. Then, we used this m-f-PBPK to: 1) perform a series of sensitivity analyses to quantitatively demonstrate the impact of fetoplacental metabolism and placental transport on fetal drug exposure for various drug-dosing regimens administered to the mother; 2) predict the impact of gestational age on fetal drug exposure; and 3) demonstrate that a single umbilical venous (UV)/maternal plasma (MP) ratio (even after multiple-dose oral administration to steady state) does not necessarily reflect fetal drug exposure. In addition, we verified the implementation of this m-f-PBPK model by comparing the predicted UV/MP and fetal/MP AUC ratios with those predicted at steady state after an intravenous infusion. Our simulations yielded novel insights into the quantitative contribution of fetoplacental metabolism and/or placental transport on gestational age-dependent fetal drug exposure. Through sensitivity analyses, we demonstrated that the UV/MP ratio does not measure the extent of fetal drug exposure unless obtained at steady state after an intravenous infusion or when there is little or no fluctuation in MP drug concentrations after multiple-dose oral administration. The proposed m-f-PBPK model can be used to predict fetal exposure to drugs across gestational ages and therefore provide the necessary information to assess the risk of drug toxicity to the fetus.
Collapse
Affiliation(s)
- Zufei Zhang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| | - Marjorie Z Imperial
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| | - Gabriela I Patilea-Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| | - Janak Wedagedera
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| | - Lu Gaohua
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (Z.Z., M.Z.I., G.I.P.-V, J.D.U.); and Simcyp Limited (a Certara company), Sheffield, United Kingdom (J.W., L.G.)
| |
Collapse
|
164
|
Tanaka S, Kanagawa T, Momma K, Hori S, Satoh H, Nagamatsu T, Fujii T, Kimura T, Sawada Y. Prediction of sustained fetal toxicity induced by ketoprofen based on PK/PD analysis using human placental perfusion and rat toxicity data. Br J Clin Pharmacol 2017. [PMID: 28635050 DOI: 10.1111/bcp.13352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIM We encountered a case of fetal toxicity due to ductus arteriosus (DA) constriction in a 36-week pregnant woman who had applied multiple ketoprofen patches. The aim of the present study was to present the case and develop a model to predict quantitatively the fetal toxicity risk of transdermal administration of ketoprofen. METHODS Human placenta perfusion studies were conducted to estimate transplacental pharmacokinetic (PK) parameters. Using a developed model and these parameters, human fetal plasma concentration profiles of ketoprofen administered to mothers were simulated. Using pregnant rats, DA constriction and fetal plasma drug concentration after ketoprofen administration were measured, fitted to an Emax model, and extrapolated to humans. RESULTS Transplacental transfer value at the steady state of ketoprofen was 4.82%, which was approximately half that of antipyrine (passive marker). The model and PK parameters predicted almost equivalent mother and fetus drug concentrations at steady state after transdermal ketoprofen administration in humans. Maximum DA constriction and maximum plasma concentration of ketoprofen after administration to rat dams were observed at different times: 4 h and 1 h, respectively. The model accurately described the delay in DA constriction with respect to the fetal ketoprofen concentration profile. The model with effect compartment and the obtained parameters predicted that use of multiple ketoprofen patches could potentially cause severe DA constriction in the human fetus, and that fetal toxicity might persist after ketoprofen discontinuation by the mother, as observed in our case. CONCLUSION The present approach successfully described the sustained fetal toxicity after discontinuing the transdermal administration of ketoprofen.
Collapse
Affiliation(s)
- Shingo Tanaka
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Takeshi Kanagawa
- Department of Maternal-Fetal Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Kazuo Momma
- Department of Pediatric Cardiology, Tokyo Women's Medical University Hospital, Tokyo, Japan
| | - Satoko Hori
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Hiroki Satoh
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasufumi Sawada
- Graduate School of Pharmaceutical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
165
|
Bérard A, Gaedigk A, Sheehy O, Chambers C, Roth M, Bozzo P, Johnson D, Kao K, Lavigne S, Wolfe L, Quinn D, Dieter K, Zhao JP. Association between CYP2D6 Genotypes and the Risk of Antidepressant Discontinuation, Dosage Modification and the Occurrence of Maternal Depression during Pregnancy. Front Pharmacol 2017; 8:402. [PMID: 28769788 PMCID: PMC5511844 DOI: 10.3389/fphar.2017.00402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 06/07/2017] [Indexed: 12/11/2022] Open
Abstract
Importance: Polymorphic expression of drug metabolizing enzymes affects the metabolism of antidepressants, and thus can contribute to drug response and/or adverse events. Pregnancy itself can affect CYP2D6 activity with profound variations determined by CYP2D6 genotype. Objective: To investigate the association between CYP2D6 genotype and the risk of antidepressant discontinuation, dosage modification, and the occurrence of maternal CYP2D6, Antidepressants, Depression during pregnancy. Setting: Data from the Organization of Teratology Information Specialists (OTIS) Antidepressants in Pregnancy Cohort, 2006-2010, were used. Women were eligible if they were within 14 completed weeks of pregnancy at recruitment and exposed to an antidepressant or having any exposures considered non-teratogenic. Main Outcomes and Measures: Gestational antidepressant usage was self-reported and defined as continuous/discontinued use, and non-use; dosage modification was further documented. Maternal depression and anxiety were measured every trimester using the telephone interviewer-administered Edinburgh Postnatal Depression Scale and the Beck Anxiety Inventory, respectively. Saliva samples were collected and used for CYP2D6 genotype analyses. Logistic regression models were used to calculate crude and adjusted odds ratios (OR) with 95% confidence intervals. Results: A total of 246 pregnant women were included in the study. The majority were normal metabolizers (NM, n = 204, 83%); 3.3% (n = 8) were ultrarapid metabolizers (UM), 5.7% (n = 14) poor metabolizers (PM), and 8.1% (n = 20) intermediate metabolizers (IM). Among study subjects, 139 women were treated with antidepressants at the beginning of pregnancy, and 21 antidepressant users (15%) discontinued therapy during pregnancy. Adjusting for depressive symptoms, and other potential confounders, the risk of discontinuing antidepressants during pregnancy was nearly four times higher in slow metabolizers (poor or intermediate metabolizers) compared to those with a faster metabolism rate (normal or ultrarapid metabolizers), aOR = 3.57 (95% CI: 1.15-11.11). Predicted CYP2D6 metabolizer status did not impact dosage modifications. Compared with slow metabolizers, significantly higher proportion of women in the fast metabolizer group had depressive symptom in the first trimester (19.81 vs. 5.88%, P = 0.049). Almost 21% of treated women remained depressed during pregnancy (14.4% NM-UM; 6.1% PM-IM). Conclusions and Relevance: Prior knowledge of CYP2D6 genotype may help to identify pregnant women at greater risk of antidepressant discontinuation. Twenty percent of women exposed to antidepressants during pregnancy remained depressed, indicating an urgent need for personalized treatment of depression during pregnancy.
Collapse
Affiliation(s)
- Anick Bérard
- Faculty of Pharmacy, University of MontrealMontreal, QC, Canada
- Research Center, CHU Sainte-JustineMontreal, QC, Canada
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy-Kansas CityKansas City, MO, United States
- School of Medicine, University of Missouri-Kansas CityKansas City, MO, United States
| | - Odile Sheehy
- Research Center, CHU Sainte-JustineMontreal, QC, Canada
| | - Christina Chambers
- Department of Pediatrics, University of California San DiegoLa Jolla, CA, United States
| | - Mark Roth
- Pregnancy Risk Network, NYS Teratogen Information ServiceBinghamton, NY, United States
| | - Pina Bozzo
- Motherisk Program, Hospital for Sick ChildrenToronto, ON, Canada
| | - Diana Johnson
- California Teratogen Information ServiceSan Diego, CA, United States
| | - Kelly Kao
- California Teratogen Information ServiceSan Diego, CA, United States
| | - Sharon Lavigne
- Connecticut Pregnancy Exposure Information Service, Division of Human Genetics, University of Connecticut Health CenterFarmington, CT, United States
| | - Lori Wolfe
- Texas Teratogen Information Service, University of North TexasDenton, TX, United States
| | - Dee Quinn
- Arizona Pregnancy Riskline, Colleges of Medicine and Pharmacy, University of ArizonaTucson, AZ, United States
| | - Kristen Dieter
- Illinois Teratology Information ServiceChicago, IL, United States
| | - Jin-Ping Zhao
- Research Center, CHU Sainte-JustineMontreal, QC, Canada
| | | |
Collapse
|
166
|
Westin AA, Brekke M, Molden E, Skogvoll E, Spigset O. Selective serotonin reuptake inhibitors and venlafaxine in pregnancy: Changes in drug disposition. PLoS One 2017; 12:e0181082. [PMID: 28708853 PMCID: PMC5510868 DOI: 10.1371/journal.pone.0181082] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/26/2017] [Indexed: 01/24/2023] Open
Abstract
Background Pregnancy may cause changes in drug disposition. The clinical consequences may be profound and even counterintuitive; in some cases pregnant women may need more than twice their usual drug dose in order to maintain therapeutic drug levels. For antidepressants, evidence on drug disposition in pregnancy is scarce. The aim of this study was to determine the effects of pregnancy on serum levels of selective serotonin reuptake inhibitors (SSRIs) and venlafaxine in a large and naturalistic patient material, in order to provide tentative dose recommendations for pregnant women. Methods Using patient data from two routine therapeutic drug monitoring (TDM) services in Norway with linkage to the national birth registry, dose-adjusted serum drug concentrations of SSRIs and venlafaxine during pregnancy were compared to the women’s own baseline (non-pregnant) values, using a linear mixed model. Findings Overall, the TDM databases contained 196,726 serum concentration measurements from 54,393 women. After data linkage and drug selection (SSRIs or venlafaxine only), we identified 367 analyses obtained from a total of 290 pregnancies in 281 women, and 420 baseline observations from the same women. Serum concentrations in the third trimester were significantly lower than baseline for paroxetine (–51%; 95% confidence interval [CI], –66%, –30%; p<0.001), fluvoxamine (–56%; CI, –75%, –23%; p = 0.004) and citalopram (–24%; CI, –38%, –7%; p = 0,007), and higher than baseline for sertraline (+68%; CI, +37%, +106%; p<0.001). For escitalopram, fluoxetine and venlafaxine concentrations did not change significantly. Conclusions For paroxetine and fluvoxamine the pronounced decline in maternal drug serum concentrations in pregnancy may necessitate a dose increase of about 100% during the third trimester in order to maintain stable concentrations. For fluoxetine, venlafaxine, citalopram, escitalopram and sertraline, the present study indicates that dose adjustments are generally not necessary during pregnancy.
Collapse
Affiliation(s)
| | - Malin Brekke
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Eirik Skogvoll
- Department of Anaesthesiology and Intensive Care, St. Olav University Hospital, Trondheim, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Olav Spigset
- Department of Clinical Pharmacology, St Olav University Hospital, Trondheim, Norway
- Department of Laboratory Medicine, Children’s and Women’s Health, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
167
|
Jogiraju VK, Avvari S, Gollen R, Taft DR. Application of physiologically based pharmacokinetic modeling to predict drug disposition in pregnant populations. Biopharm Drug Dispos 2017; 38:426-438. [DOI: 10.1002/bdd.2081] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Vamshi Krishna Jogiraju
- Samuel J. and Joan B. Williamson Institute for Pharmacometrics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences; Long Island University; Brooklyn New York USA
| | - Suvarchala Avvari
- Samuel J. and Joan B. Williamson Institute for Pharmacometrics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences; Long Island University; Brooklyn New York USA
| | - Rakesh Gollen
- Samuel J. and Joan B. Williamson Institute for Pharmacometrics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences; Long Island University; Brooklyn New York USA
- KinderPharm LLC; Exton Pennsylvania USA
| | - David R. Taft
- Samuel J. and Joan B. Williamson Institute for Pharmacometrics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences; Long Island University; Brooklyn New York USA
| |
Collapse
|
168
|
Thornton S, Valenzuela G, Baidoo C, Fossler MJ, Montague TH, Clayton L, Powell M, Snidow J, Stier B, Soergel D. Treatment of spontaneous preterm labour with retosiban: a phase II pilot dose-ranging study. Br J Clin Pharmacol 2017; 83:2283-2291. [PMID: 28556962 PMCID: PMC5595955 DOI: 10.1111/bcp.13336] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 05/12/2017] [Accepted: 05/17/2017] [Indexed: 11/27/2022] Open
Abstract
Aims The aims of the present study were to investigate the maternal, fetal and neonatal safety and tolerability, pharmacodynamics and pharmacokinetics of intravenous (IV) retosiban in pregnant women with spontaneous preterm labour (PTL) between 340/7 and 356/7 weeks' gestation. Methods In parts A and B of a three‐part, double‐blind, placebo‐controlled, multicentre study, women were randomized 3:1 (Part A) or 2:1 (Part B) to either 12‐h IV retosiban followed by a single dose of oral placebo (R‐P) or 12‐h IV placebo followed by single‐dose oral retosiban (P‐R). Results A total of 29 women were randomized; 20 to R‐P and nine to P‐R. An integrated analysis found that adverse events were infrequent in mothers/newborns and consistent with events expected in the population under study or associated with confounding factors. Retosiban was rapidly absorbed after oral administration, with an observed half‐life of 1.45 h. Efficacy analyses included 19 women. While not statistically significant, those receiving R‐P more frequently achieved uterine quiescence in 6 h (R‐P, 63%; 95% credible interval [CrI]: 38, 84; P‐R, 43%; 95% CrI: 12, 78) and more achieved a reduction of ≥50% in uterine contractions in 6 h (R‐P, 63%; 95% CrI: 38, 84; P‐R, 29%; 95% CrI: 4, 64). The number of days to delivery was increased in women receiving R‐P (median 26 days for R‐P vs. 13 days for P‐R). Conclusions Intravenous retosiban has a favourable safety and tolerability profile and might prolong pregnancies in women with PTL. The study provides the rationale and dosing strategy for further evaluation of the efficacy of retosiban in the treatment of PTL.
Collapse
Affiliation(s)
- Steven Thornton
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | - Michael J Fossler
- Quantitative Sciences, GSK, Uxbridge, UK.,Quantitative Sciences, GSK, King of Prussia, PA, USA
| | - Timothy H Montague
- Quantitative Sciences, GSK, Uxbridge, UK.,Quantitative Sciences, GSK, King of Prussia, PA, USA
| | | | | | | | | | | |
Collapse
|
169
|
Mutagonda RF, Kamuhabwa AAR, Minzi OMS, Massawe SN, Asghar M, Homann MV, Färnert A, Aklillu E. Effect of pharmacogenetics on plasma lumefantrine pharmacokinetics and malaria treatment outcome in pregnant women. Malar J 2017; 16:267. [PMID: 28673292 PMCID: PMC5496343 DOI: 10.1186/s12936-017-1914-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 06/27/2017] [Indexed: 01/09/2023] Open
Abstract
Background Pregnancy has considerable effects on the pharmacokinetic properties of drugs used to treat uncomplicated Plasmodium falciparum malaria. The role of pharmacogenetic variation on anti-malarial drug disposition and efficacy during pregnancy is not well investigated. The study aimed to examine the effect of pharmacogenetics on lumefantrine (LF) pharmacokinetics and treatment outcome in pregnant women. Methods Pregnant women with uncomplicated falciparum malaria were enrolled and treated with artemether-lumefantrine (ALu) at Mkuranga and Kisarawe district hospitals in Coast Region of Tanzania. Day-7 LF plasma concentration and genotyping forCYP2B6 (c.516G>T, c.983T>C), CYP3A4*1B, CYP3A5 (*3, *6, *7) and ABCB1 c.4036A4G were determined. Blood smear for parasite quantification by microscopy, and dried blood spot for parasite screening and genotyping using qPCR and nested PCR were collected at enrolment up to day 28 to differentiate between reinfection from recrudescence. Treatment response was recorded following the WHO protocol. Results In total, 92 pregnant women in their second and third trimester were included in the study and 424 samples were screened for presence of P. falciparum. Parasites were detected during the follow up period in 11 (12%) women between day 7 and 28 after treatment and PCR genotyping confirmed recrudescent infection in 7 (63.3%) women. The remaining four (36.4%) pregnant women had reinfection: one on day 14 and three on day 28. The overall PCR-corrected treatment failure rate was 9.0% (95% CI 4.4–17.4). Day 7 LF concentration was not significantly influenced by CYP2B6, CYP3A4*1B and ABCB1 c.4036A>G genotypes. Significant associations between CYP3A5 genotype and day 7 plasma LF concentrations was found, being higher in carriers of CYP3A5 defective variant alleles than CYP3A5*1/*1 genotype. No significant influence of CYP2B6, CYP3A5 and ABCB1 c.4036A>Genotypes on malaria treatment outcome were observed. However, CYP3A4*1B did affect malaria treatment outcome in pregnant women followed up for 28 days (P = 0.018). Conclusions Genetic variations in CYP3A4 and CYP3A5may influence LF pharmacokinetics and treatment outcome in pregnant women.
Collapse
Affiliation(s)
- Ritah F Mutagonda
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, P.O. Box 65013, Dar es Salaam, Tanzania.
| | - Appolinary A R Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, P.O. Box 65013, Dar es Salaam, Tanzania
| | - Omary M S Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, P.O. Box 65013, Dar es Salaam, Tanzania
| | - Siriel N Massawe
- Department of Obstetrics and Gynaecology, School of Medicine, Muhimbili University of Allied Sciences, P.O Box 65013, Dar es Salaam, Tanzania
| | - Muhammad Asghar
- Unit of Infectious Diseases, Department of Medicine, Karolinska Institutet, Solna, 171 76, Stockholm, Sweden
| | - Manijeh V Homann
- Unit of Infectious Diseases, Department of Medicine, Karolinska Institutet, Solna, 171 76, Stockholm, Sweden
| | - Anna Färnert
- Unit of Infectious Diseases, Department of Medicine, Karolinska Institutet, Solna, 171 76, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Eleni Aklillu
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
170
|
Leffert LR, Dubois HM, Butwick AJ, Carvalho B, Houle TT, Landau R. Neuraxial Anesthesia in Obstetric Patients Receiving Thromboprophylaxis With Unfractionated or Low-Molecular-Weight Heparin: A Systematic Review of Spinal Epidural Hematoma. Anesth Analg 2017. [DOI: 10.1213/ane.0000000000002173] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
171
|
Salman S, Baiwog F, Page-Sharp M, Kose K, Karunajeewa HA, Mueller I, Rogerson SJ, Siba PM, Ilett KF, Davis TME. Optimal antimalarial dose regimens for chloroquine in pregnancy based on population pharmacokinetic modelling. Int J Antimicrob Agents 2017; 50:542-551. [PMID: 28669839 DOI: 10.1016/j.ijantimicag.2017.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/25/2022]
Abstract
Despite extensive use and accumulated evidence of safety, there have been few pharmacokinetic studies from which appropriate chloroquine (CQ) dosing regimens could be developed specifically for pregnant women. Such optimised CQ-based regimens, used as treatment for acute malaria or as intermittent preventive treatment in pregnancy (IPTp), may have a valuable role if parasite CQ sensitivity returns following reduced drug pressure. In this study, population pharmacokinetic/pharmacodynamic modelling was used to simultaneously analyse plasma concentration-time data for CQ and its active metabolite desethylchloroquine (DCQ) in 44 non-pregnant and 45 pregnant Papua New Guinean women treated with CQ and sulfadoxine/pyrimethamine or azithromycin (AZM). Pregnancy was associated with 16% and 49% increases in CQ and DCQ clearance, respectively, as well as a 24% reduction in CQ relative bioavailability. Clearance of DCQ was 22% lower in those who received AZM in both groups. Simulations based on the final multicompartmental model demonstrated that a 33% CQ dose increase may be suitable for acute treatment for malaria in pregnancy as it resulted in equivalent exposure to that in non-pregnant women receiving recommended doses, whilst a double dose would likely be required for an effective duration of post-treatment prophylaxis when used as IPTp especially in areas of CQ resistance. The impact of co-administered AZM was clinically insignificant in simulations. The results of past/ongoing trials employing recommended adult doses of CQ-based regimens in pregnant women should be interpreted in light of these findings, and consideration should be given to using increased doses in future trials.
Collapse
Affiliation(s)
- Sam Salman
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Francesca Baiwog
- Papua New Guinea Institute of Medical Research, P.O. Box 378, Madang, Madang Province, Papua New Guinea
| | - Madhu Page-Sharp
- School of Pharmacy, Curtin University of Technology, Kent Street, Bentley, WA 6102, Australia
| | - Kay Kose
- Papua New Guinea Institute of Medical Research, P.O. Box 378, Madang, Madang Province, Papua New Guinea
| | - Harin A Karunajeewa
- Population Health and Immunity, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Ivo Mueller
- Population Health and Immunity, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3052, Australia; Barcelona Institute for Global Health (ISGlobal), Carrer del Rosselló 132, 08036 Barcelona, Spain
| | - Stephen J Rogerson
- Department of Medicine (RMH), The University of Melbourne, 300 Grattan Street, Parkville, VIC 3050, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, P.O. Box 378, Madang, Madang Province, Papua New Guinea
| | - Kenneth F Ilett
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Timothy M E Davis
- School of Medicine and Pharmacology, University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia.
| |
Collapse
|
172
|
Pharmacokinetics of Total and Unbound Etravirine in HIV-1-Infected Pregnant Women. J Acquir Immune Defic Syndr 2017; 73:268-274. [PMID: 27159225 DOI: 10.1097/qai.0000000000001068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Treatment of HIV-1-infected women during pregnancy protects maternal health and reduces the risk of perinatal transmission of HIV-1. However, physiologic changes that occur during pregnancy may affect drug pharmacokinetics. This phase IIIb, open-label study evaluated the effects of pregnancy on the pharmacokinetics of the nonnucleoside reverse transcriptase inhibitor etravirine. METHODS Eligible HIV-1-infected pregnant women (18-26 weeks gestation) on an individualized, highly active antiretroviral therapy regimen including etravirine 200 mg twice daily were enrolled. Blood samples to assess the pharmacokinetics of total and unbound etravirine were obtained at clinic visits during the second and third trimesters (24- to 28-weeks and 34- to 38-weeks gestation, respectively) and 6-12 weeks postpartum. At each time point, plasma concentrations were measured over 12 hours (12-hour time point was obtained before the second daily dose of etravirine); pharmacokinetic parameters were derived using noncompartmental analysis and were compared between pregnancy and postpartum using general linear models. Antiviral and immunologic response and safety were assessed at each visit. RESULTS Etravirine pharmacokinetic profiles were available for 13 of 15 enrolled women. Exposure to total etravirine was generally higher during pregnancy compared with 6-12 weeks postpartum (1.2- to 1.4-fold); the differences were less pronounced for unbound (pharmacodynamically active) etravirine. Virologic response was generally preserved throughout the study, and no perinatal transmission was observed. Etravirine was generally safe and well tolerated. CONCLUSIONS Etravirine 200 mg twice daily, as part of individualized combination antiretroviral therapy, may be a treatment option for HIV-1-infected pregnant women.
Collapse
|
173
|
Soma M, Konda A, Fujieda S, Sasaki Y, Keira M, Yoshida H, Mukai Y, Toda T, Inotsume N. Concentration of Sulfate and Glucuronide Conjugates of Ritodrine in Twin Pregnancy. Biol Pharm Bull 2017; 40:922-925. [PMID: 28566635 DOI: 10.1248/bpb.b16-00818] [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: 11/22/2022]
Abstract
Ritodrine, a drug for the treatment of threatened premature labor, is a highly selective beta-2 agonist with the major metabolites of sulfate and glucuronide conjugates. This study investigated the continuous evaluation of the concentration of ritodrine conjugates in relation to the clinical course in twin pregnancy. The subjects were 9 twin-pregnancy mothers who delivered after receiving ritodrine treatment between April 2012 and December 2013. Serum ritodrine sulfate and glucuronide conjugates were deconjugated using their specific enzymes. Ritodrine concentration was measured by liquid chromatography-tandem mass spectrometry. The continuous infusion rate of ritodrine was 2.66±0.67 (0.8-3.54) µg/min/kg, and the average concentration of unchanged ritodrine was 118.8±33.2 (63.8-194.0) ng/mL. During the study period between week 32 and week 36 of gestation, the average ratio of unchanged ritodrine concentration and sulfate ritodrine conjugate concentration for weeks 32, 33, 34, 35, and 36 were 1.7, 1.9, 1.5, 1.7, and 1.7 not significant (N.S.), respectively. The average ratio of unchanged ritodrine concentration and glucuronide ritodrine conjugate concentration were 1.8, 2.2, 1.9, 1.8, and 2.1 (N.S.), respectively. No statistical difference was identified in the ratios of unchanged ritodrine concentration and sulfate or glucuronide ritodrine conjugate concentrations. Large individual differences were shown in the concentration of sulfate and glucuronide during the gestational period. No change in the ratio of the formation of ritodrine metabolites was identified as the gestational age progressed.
Collapse
Affiliation(s)
| | - Ainari Konda
- Hokkaido Pharmaceutical University School of Pharmacy
| | - Satoko Fujieda
- Department of Obstetrics and Gynecology, Tenshi Hospital
| | | | - Mitsuaki Keira
- Department of Obstetrics and Gynecology, Tenshi Hospital
| | | | - Yuji Mukai
- Hokkaido Pharmaceutical University School of Pharmacy
| | - Takaki Toda
- Hokkaido Pharmaceutical University School of Pharmacy
| | | |
Collapse
|
174
|
Ragni MV, Machin N, James AH, Seaman CD, Malec LM, Kessler CM, Konkle BA, Kouides PA, Neff AT, Philipp CS, Brooks MM. Feasibility of the Von Willebrand disease PREVENT trial. Thromb Res 2017; 156:8-13. [PMID: 28577390 DOI: 10.1016/j.thromres.2017.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/20/2017] [Accepted: 05/23/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Despite treatment, women with von Willebrand disease (VWD) have lower von Willebrand factor (VWF) levels and greater blood loss at delivery than controls. Current weight-based dosing does not account for the ~1.5-fold increase in blood volume in pregnancy. METHODS To evaluate the feasibility of a trial to prevent postpartum hemorrhage (PPH), we reviewed pre-pregnancy and 8th month VWF levels in women with VWD with and without PPH following vaginal delivery, assessed VWF concentrate use at delivery by U.S. hemophilia treatment center physician survey, and reviewed thrombosis risk with VWF concentrate by literature review. We determined trial interest and acceptability by structured interviews of physicians and patients. Analysis was by Student's t-test for continuous data, and chi-square or Fisher's exact test for discrete data. RESULTS PPH was associated with lower pre-pregnancy VWF:RCo, p<0.005; higher pre-pregnancy, 8th and 9th-month weight, each p<0.001; a family bleeding history, p=0.036; and VWF concentrate treatment, p=0.005. Surveyed physicians reported first-line therapy at delivery was VWF concentrate, at a mean dose 50IU/kg. A trial of a 1.5-fold volume-based dose increase was acceptable to physicians and patients, if it is safe and if costs and visits are minimized. A literature review determined thrombosis risk with VWF concentrate is low, 0.4%. CONCLUSIONS This study suggests pre-pregnancy VWF:RCo may predict PPH, but 50-80IU/kg VWF concentrate dosing may not prevent PPH. If pharmacokinetic modeling confirms volume-based dosing achieves VWF levels comparable to pregnant controls, it may be possible to determine if volume-modified VWF concentrate dosing will reduce PPH in VWD.
Collapse
Affiliation(s)
- Margaret V Ragni
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States; Hemophilia Center of Western Pennsylvania, Pittsburgh, PA, United States.
| | - Nicoletta Machin
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Andra H James
- Duke University Medical Center, Durham, NC, United States
| | - Craig D Seaman
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States; Hemophilia Center of Western Pennsylvania, Pittsburgh, PA, United States
| | - Lynn M Malec
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States; Hemophilia Center of Western Pennsylvania, Pittsburgh, PA, United States
| | - Craig M Kessler
- Georgetown University Medical Center, Washington, DC, United States
| | | | | | - Anne T Neff
- Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Claire S Philipp
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Maria M Brooks
- Graduate School of Public Health, Pittsburgh, PA, United States
| |
Collapse
|
175
|
Välitalo PA, Kemppainen H, Kulo A, Smits A, van Calsteren K, Olkkola KT, de Hoon J, Knibbe CAJ, Allegaert K. Body weight, gender and pregnancy affect enantiomer-specific ketorolac pharmacokinetics. Br J Clin Pharmacol 2017; 83:1966-1975. [PMID: 28429492 DOI: 10.1111/bcp.13311] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/23/2017] [Accepted: 04/15/2017] [Indexed: 12/20/2022] Open
Abstract
AIMS Although ketorolac analgesia is linked only to the S-enantiomer, there is limited information on the stereo-selective pharmacokinetics of this agent. We studied the stereo-selective pharmacokinetics of ketorolac in a pooled dataset of two studies, with women at delivery and 4-5 months postpartum, and males and nonpregnant females. METHODS Nonlinear mixed-effect modelling was used to evaluate the stereo-selective pharmacokinetics of ketorolac tromethamine after a single intravenous injection immediately after delivery (n = 41), 4-5 months postpartum (n = 8, paired), and in male (n = 12) and nonpregnant female (n = 14) subjects. All of the males and six of the nonpregnant females were recruited from another study, in which they were undergoing blood sampling for 24 h. All remaining cases underwent blood sampling for 8 h. RESULTS For both the R- and S-enantiomers, body weight affected ketorolac clearance. In addition, clearance for both enantiomers was 36% [95% confidence interval (CI) 15%, 58%] higher in male than in female subjects of the same body weight, and 55% (95% CI 33%, 78%) higher in women at delivery than in nonpregnant women of the same body weight. Women at delivery also had a 27% (95% CI 8%, 46%) higher distribution volume than nonpregnant women. The proportional effects of the covariates were not significantly different for the two ketorolac enantiomers. CONCLUSIONS Besides the anticipated impact of body weight on clearance, R- and S-ketorolac clearance is increased in male subjects and in women at delivery. To reach an exposure equivalent to that in nonpregnant women, males should receive a 36% increased ketorolac dose and pregnant women a 55% increased dose, in addition to a dose adjustment by body weight.
Collapse
Affiliation(s)
- Pyry A Välitalo
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Heidi Kemppainen
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.,School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Aida Kulo
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Anne Smits
- Neonatal Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Kristel van Calsteren
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Klaus T Olkkola
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jan de Hoon
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium.,Center for Clinical Pharmacology, University Hospitals Leuven, Leuven, Belgium
| | - Catherijne A J Knibbe
- Division of Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.,Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, the Netherlands
| | - Karel Allegaert
- Department of Development and Regeneration, University Hospitals Leuven, Leuven, Belgium.,Department of Pediatric Surgery and Intensive Care, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| |
Collapse
|
176
|
Abstract
Pregnancy is accompanied by various physiological and physical changes, including those found in the cardiovascular, respiratory, gastrointestinal, renal and haematological systems. These alterations in the pregnant patient may potentially affect drug pharmacokinetics. Also, pharmacotherapy presents a unique matter due to the potential teratogenic effects of certain drugs. Although medications prescribed by dentists are generally safe during pregnancy, some modifications may be needed. In this article we will discuss the changes in the physiology during pregnancy and its impact on drug therapy. Specific emphasis will be given to the drugs commonly given by dentists, namely, local anaesthetics, analgesics, antibiotics and sedatives.
Collapse
|
177
|
Salman S, Baiwog F, Page-Sharp M, Griffin S, Karunajeewa HA, Mueller I, Rogerson SJ, Siba PM, Ilett KF, Davis TME. Optimal Antimalarial Dose Regimens for Sulfadoxine-Pyrimethamine with or without Azithromycin in Pregnancy Based on Population Pharmacokinetic Modeling. Antimicrob Agents Chemother 2017; 61:e02291-16. [PMID: 28242669 PMCID: PMC5404578 DOI: 10.1128/aac.02291-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/20/2017] [Indexed: 01/19/2023] Open
Abstract
Optimal dosing of sulfadoxine-pyrimethamine (SP) as intermittent preventive treatment in pregnancy remains to be established, particularly when coadministered with azithromycin (AZI). To further characterize SP pharmacokinetics in pregnancy, plasma concentration-time data from 45 nonpregnant and 45 pregnant women treated with SP-AZI (n = 15 in each group) and SP-chloroquine (n = 30 in each group) were analyzed. Population nonlinear mixed-effect pharmacokinetic models were developed for pyrimethamine (PYR), sulfadoxine (SDOX), and N-acetylsulfadoxine (the SDOX metabolite NASDOX), and potential covariates were included. Pregnancy increased the relative clearance (CL/F) of PYR, SDOX, and NASDOX by 48, 29, and 70%, respectively, as well as the relative volumes of distribution (V/F) of PYR (46 and 99%) and NASDOX (46%). Coadministration of AZI resulted in a greater increase in PYR CL/F (80%) and also increased NASDOX V/F by 76%. Apparent differences between these results and those of published studies of SP disposition may reflect key differences in study design, including the use of an early postpartum follow-up study rather than a nonpregnant comparator group. Simulations based on the final population model demonstrated that, compared to conventional single-dose SP in nonpregnant women, two such doses given 24 h apart should ensure that pregnant women have similar drug exposure, while three daily SP doses may be required if SP is given with AZI. The results of past and ongoing trials using recommended adult SP doses with or without AZI in pregnant women may need to be interpreted in light of these findings and consideration given to using increased doses in future trials.
Collapse
Affiliation(s)
- Sam Salman
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Francisca Baiwog
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Madhu Page-Sharp
- School of Pharmacy, Curtin University of Technology, Bentley, Western Australia, Australia
| | - Susan Griffin
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Harin A Karunajeewa
- Population Health and Immunity, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
| | - Ivo Mueller
- Population Health and Immunity, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Barcelona Institute for Global Health (ISGLOBAL), Barcelona, Spain
| | - Stephen J Rogerson
- Department of Medicine (RMH), The University of Melbourne, Parkville, Victoria, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Kenneth F Ilett
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Timothy M E Davis
- School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
178
|
Blood volume-based von Willebrand factor to prevent postpartum hemorrhage in von Willebrand disease. Blood Adv 2017; 1:703-706. [PMID: 29296713 DOI: 10.1182/bloodadvances.2017005090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Publisher's Note: This article has a companion Point by Kouides. Publisher's Note: Join in the discussion of these articles at Blood Advances Community Conversations.
Collapse
|
179
|
Bookstaver PB, Bland CM, Griffin B, Stover KR, Eiland LS, McLaughlin M. A Review of Antibiotic Use in Pregnancy. Pharmacotherapy 2017; 35:1052-62. [PMID: 26598097 DOI: 10.1002/phar.1649] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During pregnancy, untreated sexually transmitted or urinary tract infections are associated with significant morbidity, including low birth weight, preterm birth, and spontaneous abortion. Approximately one in four women will be prescribed an antibiotic during pregnancy, accounting for nearly 80% of prescription medications in pregnant women. Antibiotic exposures during pregnancy have been associated with both short-term (e.g., congenital abnormalities) and long-term effects (e.g., changes in gut microbiome, asthma, atopic dermatitis) in the newborn. However, it is estimated that only 10% of medications have sufficient data related to safe and effective use in pregnancy. Antibiotics such as beta-lactams, vancomycin, nitrofurantoin, metronidazole, clindamycin, and fosfomycin are generally considered safe and effective in pregnancy. Fluoroquinolones and tetracyclines are generally avoided in pregnancy. Physiologic changes in pregnancy lead to an increase in glomerular filtration rate, increase in total body volume, and enhanced cardiac output. These changes may lead to pharmacokinetic alterations in antibiotics that require dose adjustment or careful monitoring and assessment.
Collapse
Affiliation(s)
- P Brandon Bookstaver
- Department of Clinical Pharmacy and Outcomes Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina
| | - Christopher M Bland
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Savannah, Georgia
| | - Brooke Griffin
- Department of Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| | - Kayla R Stover
- Department of Pharmacy Practice, University of Mississippi School of Pharmacy, Jackson, Mississippi
| | - Lea S Eiland
- Department of Pharmacy Practice, Auburn University Harrison School of Pharmacy, Meridian, Mississippi
| | - Milena McLaughlin
- Department of Pharmacy Practice, Midwestern University Chicago College of Pharmacy, Downers Grove, Illinois
| |
Collapse
|
180
|
Deshpande PO, Mohan V, Pore MP, Gumaste S, Thakurdesai PA. Prenatal Developmental Toxicity Study of Glycosides-based Standardized Fenugreek Seed Extract in Rats. Pharmacogn Mag 2017; 13:S135-S141. [PMID: 28479738 PMCID: PMC5407105 DOI: 10.4103/0973-1296.203978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/03/2016] [Indexed: 11/12/2022] Open
Abstract
Context: Glycoside-based standardized fenugreek seed extract (SFSE-G) demonstrated promising efficacy in animal models of immune-inflammatory conditions. Aim: The present study was aimed at embryo-fetal development toxicity evaluation of SFSE-G in Wistar rats as per guideline No. 414 of the Organization for Economic Co-operation and Development (OECD). Material and Methods: Mated female rats were randomized into four groups of 30 each and received oral doses of either SFSE-G at 250, 500, and 1000 mg/kg or vehicle (water) during the period of gestation (postconception) from gestational day 5 (GD5, an implantation day) until 1 day before cesarean sections (GD19). Maternal food consumption, body weights, and clinical signs were monitored throughout gestation. Cesarean sections were performed on GD20 and fetal observations (gravid uterine weight, implantation sites, early and late resorptions, live and dead fetuses) were recorded. Live fetuses were weighed and examined for external, visceral, and skeletal variations and malformations. Results: None of the SFSE-G-treated groups showed maternal and embryo–fetal toxicity. Occasional and incidental skeletal and visceral malformations were observed and found to be spontaneous and unrelated to the treatment. Conclusion: Oral exposure of SFSE-G during the prenatal period did not show significant maternal and embryo-fetal toxicity up to a dose of 1000 mg/kg in rats. Therefore, the no-observed-adverse-effect level for SFSE-G for prenatal oral exposure was considered to be 1000 mg/kg. SUMMARY Prenatal toxicity of glycoside-based standardized fenugreek seed extract (SFSE-G) was evaluated. SFSE-G was orally gavaged to rats on gestational days 5-19 with a limit dose of 1000 mg/kg. SFSE-G did not show maternal or developmental toxicity. SFSE-G showed NOAEL of 1000 mg/kg for prenatal exposure in female rats.
Abbreviations used: CPCSEA: Committee for the Purpose of Control and Supervision of Experiments on Animals; GD: Gestational day; GRAS: Generally recognized as safe; HED: Human equivalent dose; NOAEL: No-observed adverse effect levels; OECD: Organization for Economic Co-operation and Development; SFSE-G: glycoside-based standardized fenugreek seed extract.
Collapse
Affiliation(s)
- Pallavi O Deshpande
- Department of Scientific affairs, Indus Biotech Private Limited, Kondhwa, Pune, Maharashtra, India
| | - Vishwaraman Mohan
- Department of Scientific affairs, Indus Biotech Private Limited, Kondhwa, Pune, Maharashtra, India
| | - Mukul P Pore
- Intox Pvt. Ltd, Urawade, Mulshi, Pirangut, Pune, Maharashtra, India
| | - Shailesh Gumaste
- Intox Pvt. Ltd, Urawade, Mulshi, Pirangut, Pune, Maharashtra, India
| | - Prasad A Thakurdesai
- Department of Scientific affairs, Indus Biotech Private Limited, Kondhwa, Pune, Maharashtra, India
| |
Collapse
|
181
|
Della Torre S, Maggi A. Sex Differences: A Resultant of an Evolutionary Pressure? Cell Metab 2017; 25:499-505. [PMID: 28190772 DOI: 10.1016/j.cmet.2017.01.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/27/2016] [Accepted: 01/11/2017] [Indexed: 11/19/2022]
Abstract
Spurred by current research policy, we are witnessing a significant growth in the number of studies that observe and describe sexual diversities in human physiology and sex prevalence in a large number of pathologies. Yet we are far from the comprehension of the mechanisms underpinning these differences, which are the result of a long evolutionary history. This Essay is meant to underline female reproductive function as a driver for the positive selection of the specific physiological features that explain male and female differential susceptibility to diseases and metabolic disturbances, in particular. A clear understanding of the causes underlying sexual dimorphisms in the physio-pathology is crucial for precision medicine.
Collapse
Affiliation(s)
- Sara Della Torre
- Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti 9, 20133 Milan, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
182
|
Willemin ME, Lumen A. Thiocyanate: a review and evaluation of the kinetics and the modes of action for thyroid hormone perturbations. Crit Rev Toxicol 2017. [DOI: 10.1080/10408444.2017.1281590] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Marie-Emilie Willemin
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Annie Lumen
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| |
Collapse
|
183
|
Lange B, Mueller JK, Leweke FM, Bumb JM. How gender affects the pharmacotherapeutic approach to treating psychosis - a systematic review. Expert Opin Pharmacother 2017; 18:351-362. [PMID: 28129701 DOI: 10.1080/14656566.2017.1288722] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The effectiveness, effective dosages and side effect profiles of antipsychotic medication differ significantly between the sexes. Areas covered: We present a systematic review of gender-differences in the treatment of psychosis focusing on randomized, controlled trials and meta-analyses. Expert opinion: Despite many years of research, the database on gender-differences affecting the pharmacotherapeutic approach to treating psychosis is insufficient. Currently, the US National Institute of Health encouraged the enrolment of female participants in federally supported phase III clinical trials to increase the data available of female patients. Emerging evidence points to a superior antipsychotic response in women, with men requiring higher dosages. In general, women metabolize drugs differently, resulting in side effects occuring more frequently when compared to men. In any case, women require electrocardiograms or bone density scans as well as diabetes and cardiovascular workups when treated with antipsychotics. Dose adjustments during the menstrual cycle (e.g. to raise antipsychotic doses premenstrually) should be considered. First-generation antipsychotics, drugs that are known to prolong QTc interval and increase prolactin levels should be avoided in postmenopausal female patients. Furthermore, the effects of antipsychotics during pregnancy and breastfeeding have been investigated insufficiently, and more research is urgently needed.
Collapse
Affiliation(s)
- Bettina Lange
- a Department of Psychiatry and Psychotherapy , Central Institute of Mental Health , Mannheim , Germany
| | - Juliane K Mueller
- a Department of Psychiatry and Psychotherapy , Central Institute of Mental Health , Mannheim , Germany
| | - F Markus Leweke
- a Department of Psychiatry and Psychotherapy , Central Institute of Mental Health , Mannheim , Germany
| | - J Malte Bumb
- b Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health , Medical Faculty Mannheim/Heidelberg University , Mannheim , Germany
| |
Collapse
|
184
|
Cristofoletti R, Patel N, Dressman JB. Assessment of Bioequivalence of Weak Base Formulations Under Various Dosing Conditions Using Physiologically Based Pharmacokinetic Simulations in Virtual Populations. Case Examples: Ketoconazole and Posaconazole. J Pharm Sci 2017; 106:560-569. [DOI: 10.1016/j.xphs.2016.10.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/12/2016] [Accepted: 10/13/2016] [Indexed: 10/20/2022]
|
185
|
Filgueira GCDO, Filgueira OAS, Carvalho DM, Marques MP, Moisés ECD, Duarte G, Lanchote VL, Cavalli RC. Effect of type 2 diabetes mellitus on the pharmacokinetics and transplacental transfer of nifedipine in hypertensive pregnant women. Br J Clin Pharmacol 2017; 83:1571-1579. [PMID: 28042936 DOI: 10.1111/bcp.13226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/15/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022] Open
Abstract
AIMS Diabetes mellitus can inhibit cytochrome P450 3A4, an enzyme responsible for the metabolism of nifedipine, used for the treatment of hypertension in pregnant women. We aimed to assess the effect of type 2 diabetes mellitus (T2DM) on the pharmacokinetics, placental transfer and distribution of nifedipine in amniotic fluid in hypertensive pregnant women. METHODS The study was conducted in 12 hypertensive pregnant women [control group (CG)] and 10 hypertensive pregnant women with T2DM taking slow-release nifedipine (20 mg, 12/12 h). On the 34th week of gestation, serial blood samples were collected (0-12 h) after administration of the medication. At delivery, samples of maternal and fetal blood and amniotic fluid were collected for determination of nifedipine distribution in these compartments. RESULTS The median pharmacokinetic parameters of CG were: peak plasma concentration (Cmax ) 26.41 ng ml-1 , time to reach Cmax (tmax ) 1.79 h, area under the plasma concentration vs. time curve from 0-12 h (AUC0-12 ) 235.99 ng.h ml-1 , half-life (t½) 4.34 h, volume of distribution divided by bioavailability (Vd/F) 560.96 l, and ClT /F 84.77 l h-1 . The parameters for T2DM group were: Cmax 23.52 ng ml-1 , tmax 1.48 h, AUC0-12 202.23 ng.h ml-1 , t½ 5.00 h, Vd/F 609.40 l, and apparent total clearance (ClT /F) 98.94 l h-1 . The ratios of plasma concentrations of nifedipine in the umbilical vein, intervillous space and amniotic fluid to those in the maternal vein for CG and T2DM were 0.53 and 0.44, 0.78 and 0.87, respectively, with an amniotic fluid/maternal plasma ratio of 0.05 for both groups. The ratios of plasma concentrations in the umbilical artery to those in the umbilical vein were 0.82 for CG and 0.88 for T2DM. CONCLUSIONS There was no influence of T2DM on the pharmacokinetics or placental transfer of nifedipine in hypertensive women with controlled diabetes.
Collapse
Affiliation(s)
| | - Osmany Alberto Silva Filgueira
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Daniela Miarelli Carvalho
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maria Paula Marques
- Department of Clinical, Toxicologic and Bromatologic Analyses, Faculty of Pharmaceutical Sciences of Ribeirão Pre, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elaine Christine Dantas Moisés
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Geraldo Duarte
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical, Toxicologic and Bromatologic Analyses, Faculty of Pharmaceutical Sciences of Ribeirão Pre, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ricardo Carvalho Cavalli
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
186
|
Bastian JR, Chen H, Zhang H, Rothenberger S, Tarter R, English D, Venkataramanan R, Caritis SN. Dose-adjusted plasma concentrations of sublingual buprenorphine are lower during than after pregnancy. Am J Obstet Gynecol 2017; 216:64.e1-64.e7. [PMID: 27687214 PMCID: PMC5245872 DOI: 10.1016/j.ajog.2016.09.095] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/12/2016] [Accepted: 09/17/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Buprenorphine is a Food and Drug Administration-approved maintenance therapy for opioid use disorders and is increasingly being used in pregnant women with opioid use disorders as an alternative to methadone. Dosing of buprenorphine in pregnant women is based on the regimen recommended for nonpregnant females and males. Limited data are available defining the pharmacokinetic properties of sublingual buprenorphine administered during pregnancy. OBJECTIVE This study evaluated the impact of physiological changes associated with pregnancy on the pharmacokinetics of sublingual buprenorphine during and after pregnancy. STUDY DESIGN Pregnant women (n = 13), between 180/7 and 376/7 weeks' singleton gestation, receiving sublingual buprenorphine twice daily for opioid use disorders were studied. Pharmacokinetic-2 studies were performed between 18 and 25 weeks (n = 7), pharmacokinetic-3 studies were performed between 31 and 37 weeks (n = 11), and pharmacokinetic-P was performed 4-18 weeks postpartum (n = 10). On the day of the study, blood was withdrawn prior to the daily morning dose of buprenorphine and at 0.25, 0.5, 0.75, 1, 1.5, 2, 4, 8, and 12 hours after the dose. Buprenorphine plasma concentrations were analyzed by liquid chromatography tandem mass spectrometric detection. All pharmacokinetic parameters were observed or estimated using Microsoft Excel. Statistical analyses were performed to identify significant changes in study participants' buprenorphine pharmacokinetic parameter estimates over the duration of the study. Univariate linear and generalized linear mixed models were used to investigate changes in these measures over time, some of which were log transformed for normality. RESULTS Dose-normalized (plasma concentration per dose) buprenorphine plasma concentrations were significantly lower during pregnancy (pharmacokinetic-2 plus pharmacokinetic-3) than during the postpartum period (pharmacokinetic-P). Specific pharmacokinetic parameters (and level of significance) were as follows: the area under the buprenorphine plasma concentration-time curves (P < .003), maximum buprenorphine concentrations (P < .018), buprenorphine concentrations at 0 hour (P < .002), and buprenorphine concentrations at 12 hours (P < .001). None of these parameters differed significantly during pregnancy (ie, pharmacokinetic-2 vs pharmacokinetic-3). The time to maximum buprenorphine concentrations did not differ significantly between groups. CONCLUSION The dose-normalized plasma concentrations during a dosing interval and the overall exposure of buprenorphine (area under the buprenorphine plasma concentration-time curves) are lower throughout pregnancy compared with the postpartum period. This indicates an increase in apparent clearance of buprenorphine during pregnancy. These data suggest that pregnant women may need a higher dose of sublingual buprenorphine compared with postpartum individuals. The dose of buprenorphine should be assessed after delivery to maintain similar buprenorphine exposure during the postpartum period.
Collapse
Affiliation(s)
- Jaime R Bastian
- Divisions of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA.
| | - Huijun Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
| | - Hongfei Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
| | - Scott Rothenberger
- Center for Research on Healthcare Data Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Ralph Tarter
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
| | - Dennis English
- Divisions of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Raman Venkataramanan
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA
| | - Steve N Caritis
- Divisions of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
187
|
Duffield A, Sultan P, Riley ET, Carvalho B. Optimal administration of cefazolin prophylaxis for cesarean delivery. J Perinatol 2017; 37:16-20. [PMID: 28050017 DOI: 10.1038/jp.2016.210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/07/2016] [Accepted: 10/14/2016] [Indexed: 11/09/2022]
Affiliation(s)
- A Duffield
- Department of Anesthesiology, Sentara Martha Jefferson Hospital, Charlottesville, VA, USA
| | - P Sultan
- Department of Anaesthesia and Perioperative Medicine, University College London Hospital, London, UK
| | - E T Riley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - B Carvalho
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
188
|
Colbers A, Greupink R, Litjens C, Burger D, Russel FGM. Physiologically Based Modelling of Darunavir/Ritonavir Pharmacokinetics During Pregnancy. Clin Pharmacokinet 2016; 55:381-96. [PMID: 26369773 PMCID: PMC4761019 DOI: 10.1007/s40262-015-0325-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pregnant women are usually excluded from clinical trials. Physiologically based pharmacokinetic (PBPK) modelling may provide a method to predict pharmacokinetics in pregnant women, without the need to perform extensive in vivo clinical trials. Here, we used mechanistic modelling to delineate the potential impact of drug transporters on darunavir pharmacokinetics and to identify current knowledge gaps that limit accurate PBPK modelling of darunavir/ritonavir (darunavir/r) exposure in pregnancy. Simcyp (version 13.2) was used for PBPK modelling, using physicochemical and in vitro pharmacokinetic parameters of darunavir and ritonavir from the literature. The Michaelis-Menten constant (K m) and the maximum rate of metabolite formation (V max) for cytochrome P450 3A4-mediated darunavir biotransformation and inhibition by ritonavir were determined experimentally, while the contributions of hepatocyte influx and efflux transporters were assessed by sensitivity analysis. The simulations were compared with previously published clinical pharmacokinetic data. We found that use of a well-stirred liver model overestimated darunavir exposure substantially. A permeability-limited liver model, including hepatic uptake and efflux transporters and an efficient enterohepatic circulation step, resulted in an acceptable description of darunavir/r exposure. For the 600/100 mg darunavir/r twice-daily dose and the 800/100 mg once-daily dose, the estimated pharmacokinetic parameters were within a 2-fold range of the reported data. The predicted decreases in the area under the concentration-time curve (AUC) values during pregnancy for the twice- and once-daily doses were 27 and 41%, respectively, which were in line with the observed decreases of 17-22 and 33%. In conclusion, our data support a clinically relevant role of hepatic transporters in darunavir pharmacokinetics. By including them in our model, we successfully approximated the increase in darunavir exposure mediated by ritonavir co-administration and the decrease in darunavir exposure observed during pregnancy.
Collapse
Affiliation(s)
- Angela Colbers
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Rick Greupink
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Carlijn Litjens
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - David Burger
- Department of Pharmacy, Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology (149), Radboud University Medical Centre, Radboud Institute for Molecular Life Sciences, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
189
|
Abstract
BACKGROUND Women are commonly prescribed a variety of medications during pregnancy. As most organ systems are affected by the substantial anatomical and physiological changes that occur during pregnancy, it is expected that pharmacokinetics (PK) (absorption, distribution, metabolism, and excretion of drugs) would also be affected in ways that may necessitate changes in dosing schedules. The objective of this study was to systematically identify existing clinically relevant evidence on PK changes during pregnancy. METHODS AND FINDINGS Systematic searches were conducted in MEDLINE (Ovid), Embase (Ovid), Cochrane Central Register of Controlled Trials (Ovid), and Web of Science (Thomson Reuters), from database inception to August 31, 2015. An update of the search from September 1, 2015, to May 20, 2016, was performed, and relevant data were added to the present review. No language or date restrictions were applied. All publications of clinical PK studies involving a group of pregnant women with a comparison to nonpregnant participants or nonpregnant population data were eligible to be included in this review. A total of 198 studies involving 121 different medications fulfilled the inclusion criteria. In these studies, commonly investigated drug classes included antiretrovirals (54 studies), antiepileptic drugs (27 studies), antibiotics (23 studies), antimalarial drugs (22 studies), and cardiovascular drugs (17 studies). Overall, pregnancy-associated changes in PK parameters were often observed as consistent findings among many studies, particularly enhanced drug elimination and decreased exposure to total drugs (bound and unbound to plasma proteins) at a given dose. However, associated alterations in clinical responses and outcomes, or lack thereof, remain largely unknown. CONCLUSION This systematic review of pregnancy-associated PK changes identifies a significant gap between the accumulating knowledge of PK changes in pregnant women and our understanding of their clinical impact for both mother and fetus. It is essential for clinicians to be aware of these unique pregnancy-related changes in PK, and to critically examine their clinical implications.
Collapse
|
190
|
Fokina VM, Xu M, Rytting E, Abdel-Rahman SZ, West H, Oncken C, Clark SM, Ahmed MS, Hankins GDV, Nanovskaya TN. Pharmacokinetics of Bupropion and Its Pharmacologically Active Metabolites in Pregnancy. Drug Metab Dispos 2016; 44:1832-1838. [PMID: 27528039 PMCID: PMC5074472 DOI: 10.1124/dmd.116.071530] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/12/2016] [Indexed: 12/21/2022] Open
Abstract
Bupropion sustained release is used to promote smoking cessation in males and nonpregnant females. However, its efficacy as a smoking cessation aid during pregnancy is not reported. The pregnancy-associated changes in maternal physiology may alter the pharmacokinetics and pharmacodynamics of bupropion and consequently its efficacy in pregnant smokers. Therefore, the aims of this study were to determine the steady-state pharmacokinetics of bupropion during pregnancy and the effect of functional genetic variants of CYP2B6 and CYP2C19 on bupropion pharmacokinetics in pregnant women. Plasma and urine concentrations of bupropion and its metabolites hydroxybupropion (OHBUP), threohydrobupropion, and erythrohydrobupropion were determined by liquid chromatography-mass spectrometry. Subjects were genotyped for five nonsynonymous single-nucleotide polymorphisms that result in seven CYP2B6 alleles, namely *2, *3, *4, *5, *6, *7, and *9, and for CYP2C19 variants *2, *3, and *17 The present study reports that the isoform-specific effect of pregnancy on bupropion-metabolizing enzymes along with the increase of renal elimination of the drug could collectively result in a slight decrease in exposure to bupropion in pregnancy. In contrast, pregnancy-induced increase in CYP2B6-catalyzed bupropion hydroxylation did not impact the plasma levels of OHBUP, probably due to a higher rate of OHBUP glucuronidation, and renal elimination associated with pregnancy. Therefore, exposure to OHBUP, a pharmacologically active metabolite of the bupropion, appears to be similar to that of the nonpregnant state. The predicted metabolic phenotypes of CYP2B6*6 and variant alleles of CYP2C19 in pregnancy are similar to those in the nonpregnant state.
Collapse
Affiliation(s)
- Valentina M Fokina
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Meixiang Xu
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Erik Rytting
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Sherif Z Abdel-Rahman
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Holly West
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Cheryl Oncken
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Shannon M Clark
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Mahmoud S Ahmed
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Gary D V Hankins
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| | - Tatiana N Nanovskaya
- Department of Pharmacology and Toxicology (V.M.F.), Maternal-Fetal Pharmacology and Biodevelopment Laboratories, Department of Obstetrics and Gynecology (M.X., E.R., S.Z.A.-R., M.S.A., T.N.N.), and Department of Obstetrics and Gynecology (H.W., S.M.C., G.D.V.H.), University of Texas Medical Branch, Galveston, Texas; and University of Connecticut Health Center, Farmington, Connecticut (C.O.)
| |
Collapse
|
191
|
Alshawabkeh L, Economy KE, Valente AM. Anticoagulation During Pregnancy. J Am Coll Cardiol 2016; 68:1804-1813. [DOI: 10.1016/j.jacc.2016.06.076] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 12/22/2022]
|
192
|
Casale JP, Doligalski CT. Pharmacologic Considerations for Solid Organ Transplant Recipients Who Become Pregnant. Pharmacotherapy 2016; 36:971-82. [DOI: 10.1002/phar.1800] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
193
|
Abstract
INTRODUCTION The goal of this article is to review the use of rifapentine in the treatment of latent tuberculosis infection (LTBI). Controlling LTBI is an important part of the global strategy to end the spread of tuberculosis. Rifapentine's potent sterilizing effect against Mycobacterium tuberculosis combined with its long half-life make it an attractive LTBI treatment option. Areas covered: A systematic literature search of Pubmed using the terms 'rifapentine' and 'tuberculosis' was performed. Articles identified were cross-referenced for other relevant publications. The mechanisms of action and resistance, pharmacokinetic and pharmacodynamics, potential drug interactions and side effects are discussed. Expert commentary: Rifapentine in combination with isoniazid for twelve weeks is the best available option for treating latent TB in the majority of patients in the United States due to its favorable safety profile and the increased likelihood of completing therapy. Currently, rifapentine is not registered or available in other countries.
Collapse
Affiliation(s)
- Eric F Egelund
- a Department of Pharmacotherapy and Translational Research, College of Pharmacy , University of Florida , Gainesville , FL , USA.,b Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA
| | - Charles A Peloquin
- a Department of Pharmacotherapy and Translational Research, College of Pharmacy , University of Florida , Gainesville , FL , USA.,b Infectious Disease Pharmacokinetics Laboratory , University of Florida , Gainesville , FL , USA.,c Emerging Pathogens Institute , University of Florida , Gainesville , FL , USA
| |
Collapse
|
194
|
Mulligan N, Schalkwijk S, Best BM, Colbers A, Wang J, Capparelli EV, Moltó J, Stek AM, Taylor G, Smith E, Hidalgo Tenorio C, Chakhtoura N, van Kasteren M, Fletcher CV, Mirochnick M, Burger D. Etravirine Pharmacokinetics in HIV-Infected Pregnant Women. Front Pharmacol 2016; 7:239. [PMID: 27540363 PMCID: PMC4972814 DOI: 10.3389/fphar.2016.00239] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/21/2016] [Indexed: 01/05/2023] Open
Abstract
Background: The study goal was to describe etravirine pharmacokinetics during pregnancy and postpartum in HIV-infected women. Methods: IMPAACT P1026s and PANNA are on-going, non-randomized, open-label, parallel-group, multi-center phase-IV prospective studies in HIV-infected pregnant women. Intensive steady-state 12-h pharmacokinetic profiles were performed from 2nd trimester through postpartum. Etravirine was measured at two labs using validated ultra performance liquid chromatography (detection limits: 0.020 and 0.026 mcg/mL). Results: Fifteen women took etravirine 200 mg twice-daily. Etravirine AUC0–12 was higher in the 3rd trimester compared to paired postpartum data by 34% (median 8.3 vs. 5.3 mcg*h/mL, p = 0.068). Etravirine apparent oral clearance was significantly lower in the 3rd trimester of pregnancy compared to paired postpartum data by 52% (median 24 vs. 38 L/h, p = 0.025). The median ratio of cord blood to maternal plasma concentration at delivery was 0.52 (range: 0.19–4.25) and no perinatal transmission occurred. Conclusion: Etravirine apparent oral clearance is reduced and exposure increased during the third trimester of pregnancy. Based on prior dose-ranging and safety data, no dose adjustment is necessary for maternal health but the effects of etravirine in utero are unknown. Maternal health and infant outcomes should be closely monitored until further infant safety data are available. Clinical Trial registration: The IMPAACT protocol P1026s and PANNA study are registered at ClinicalTrials.gov under NCT00042289 and NCT00825929.
Collapse
Affiliation(s)
- Nikki Mulligan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Stein Schalkwijk
- Department of Pharmacy, Radboud University Medical Center Nijmegen, Netherlands
| | - Brookie M Best
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Angela Colbers
- Department of Pharmacy, Radboud University Medical Center Nijmegen, Netherlands
| | - Jiajia Wang
- Center for Biostatistics in AIDS Research, Harvard School of Public Health Boston, MA, USA
| | - Edmund V Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - José Moltó
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias I Pujol Badalona, Spain
| | - Alice M Stek
- Maternal Child and Adolescent/Adult Center, University of Southern California School of Medicine Los Angeles, CA, USA
| | - Graham Taylor
- Imperial College Healthcare National Health Service Trust London, UK
| | - Elizabeth Smith
- Maternal, Adolescent, and Pediatric Research Branch, National Institute of Allergy and Infectious Diseases Bethesda, MD, USA
| | | | - Nahida Chakhtoura
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development Bethesda, MD, USA
| | - Marjo van Kasteren
- Department of Internal Medicine, St. Elisabeth Hospital Tilburg, Netherlands
| | - Courtney V Fletcher
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center Omaha, NE, USA
| | - Mark Mirochnick
- Department of Pediatrics, Boston University School of Medicine Boston, MA, USA
| | - David Burger
- Department of Pharmacy, Radboud University Medical Center Nijmegen, Netherlands
| |
Collapse
|
195
|
|
196
|
Salim R, Nachum Z, Gavish I, Romano S, Braverman M, Garmi G. Adjusting enoxaparin dosage according to anti-FXa levels and pregnancy outcome in thrombophilic women. A randomised controlled trial. Thromb Haemost 2016; 116:687-95. [PMID: 27440463 DOI: 10.1160/th16-03-0221] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/22/2016] [Indexed: 02/03/2023]
Abstract
Women with thrombophilias and previous placenta-mediated pregnancy complications (PMPC) have an increased risk of both recurrent PMPC and venous thromboembolism (VTE) during subsequent pregnancies. We aimed to examine whether enoxaparin dose adjusted according to anti-factor Xa levels compared to a fixed dose would reduce the risk of PMPC in subsequent pregnancies. In a randomised trial, conducted at a single teaching hospital, pregnant women with thrombophilia and previous PMPC were enrolled in a 1:1 ratio to either a fixed dose of 40 mg daily enoxaparin or adjusted dose according to anti-factor Xa plasma levels. The primary outcome was a composite that included any of the following: pregnancy loss after enrollment, pre-eclampsia, birthweight <10th percentile, placental abruption, or VTE. Overall, 144 women were needed to detect a decrease of 20 % in the incidence of the composite outcome among the adjusted dose group. Between 2009 and 2015, 144 women consented; four in the fixed-dose group were excluded. Overall, 66 and 74 in the fixed- and adjusted-dose groups, respectively, were included. Demographic and obstetric characteristics were comparable. Composite outcome occurred in 12 (18.2 %) and 20 (27.0 %) women in the fixed- and adjusted-dose groups, respectively (p=0.24). Gestational age at delivery, preterm births, and birthweights were similar between the two groups. In conclusion, dose of enoxaparin adjusted according to anti-factor Xa levels compared to fixed dose, does not reduce the risk of PMPC recurrence in thrombophilic women.
Collapse
Affiliation(s)
- Raed Salim
- Raed Salim, MD, Department of Obstetrics and Gynecology, Emek Medical Center, Afula 18101, Israel, Tel.: +972 4 649 4031, E-mail:
| | | | | | | | | | | |
Collapse
|
197
|
Sharma S, Caritis S, Hankins G, Miodovnik M, Hebert MF, Mattison D, Venkataramanan R. Population pharmacokinetics of 17α-hydroxyprogesterone caproate in singleton gestation. Br J Clin Pharmacol 2016; 82:1084-93. [PMID: 27133963 DOI: 10.1111/bcp.12990] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/12/2016] [Accepted: 04/07/2016] [Indexed: 11/28/2022] Open
Abstract
AIMS 17α-hydroxyprogesterone caproate (17-OHPC) reduces the rate of preterm birth in women with a prior preterm birth. Limited data exist on the pharmacokinetics (PK) of 17-OHPC or the plasma concentrations achieved during therapy. In this study, we evaluated the population PK of 17-OHPC in pregnant subjects with singleton gestation and also evaluated intrinsic and extrinsic factors that may potentially affect 17-OHPC PK in this patient population. METHODS Sixty-one women with singleton pregnancies participated in this trial. Subjects received weekly intramuscular injections of 250 mg 17-OHPC in 1 ml castor oil from the time of enrolment (16 0/7 weeks - 20 6/7 weeks) up to 35 weeks gestation or until delivery. Blood samples were obtained between 24 and 28 weeks, between 32 and 35 weeks and over a 28-day period beyond the last injection. Maternal and/or cord blood were obtained at delivery. Data analysis was performed by nonlinear mixed effects modelling (NONMEM(®) ). RESULTS The 17-OHPC PK were best described by a model with one maternal compartment and one fetal compartment, with first-order absorption and elimination from the maternal compartment. Maternal body weight was a significant covariate for both clearance (CL/F) and volume of distribution (Vmaternal /F). The final population mean estimates were: CL/F 1797 l/d, Vmaternal /F 32 610 l and mother to cord rate constant 0.005 day(-1) . This report describes for the first time the population PK of 17-OHPC in singleton pregnancy. CONCLUSIONS The population PK study reported here represents the initial steps in understanding and optimizing 17-OHPC therapy for preventing preterm birth.
Collapse
Affiliation(s)
- Shringi Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Steve Caritis
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Magee-Women's Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Gary Hankins
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Menachem Miodovnik
- Medstar Health Research Institute, Hyatsville, MD, USA.,Georgetown-Howards University Center for Clinical and Translational Science, Washington, DC, USA
| | - Mary F Hebert
- Department of Pharmacy and Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| | - Don Mattison
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, and Risk Sciences International, Ottawa, ON, Canada
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
198
|
Moore BR, Salman S, Davis TME. Treatment regimens for pregnant women with falciparum malaria. Expert Rev Anti Infect Ther 2016; 14:691-704. [PMID: 27322015 DOI: 10.1080/14787210.2016.1202758] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION With increasing parasite drug resistance, the WHO has updated treatment recommendations for falciparum malaria including in pregnancy. This review assesses the evidence for choice of treatment for pregnant women. AREAS COVERED Relevant studies, primarily those published since 2010, were identified from reference databases and were used to identify secondary data sources. Expert commentary: WHO recommends use of intravenous artesunate for severe malaria, quinine-clindamycin for uncomplicated malaria in first trimester, and artemisinin combination therapy for uncomplicated malaria in second/third trimesters. Because fear of adverse outcomes has often excluded pregnant women from conventional drug development, available data for novel therapies are usually based on preclinical studies and cases of inadvertent exposure. Changes in antimalarial drug disposition in pregnancy have been observed but are yet to be translated into specific treatment recommendations. Such targeted regimens may become important as parasite resistance demands that drug exposure is optimized.
Collapse
Affiliation(s)
- Brioni R Moore
- a Fiona Stanley Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Perth , Australia.,b School of Pharmacy , Curtin University , Perth , Australia
| | - Sam Salman
- c Linear Clinical Research Limited, QEII Medical Centre , Nedlands , Australia.,d Fremantle Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Fremantle , Australia
| | - Timothy M E Davis
- d Fremantle Hospital Unit, School of Medicine and Pharmacology , University of Western Australia , Fremantle , Australia
| |
Collapse
|
199
|
Ilekis JV, Tsilou E, Fisher S, Abrahams VM, Soares MJ, Cross JC, Zamudio S, Illsley NP, Myatt L, Colvis C, Costantine MM, Haas DM, Sadovsky Y, Weiner C, Rytting E, Bidwell G. Placental origins of adverse pregnancy outcomes: potential molecular targets: an Executive Workshop Summary of the Eunice Kennedy Shriver National Institute of Child Health and Human Development. Am J Obstet Gynecol 2016; 215:S1-S46. [PMID: 26972897 DOI: 10.1016/j.ajog.2016.03.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 02/11/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
Abstract
Although much progress is being made in understanding the molecular pathways in the placenta that are involved in the pathophysiology of pregnancy-related disorders, a significant gap exists in the utilization of this information for the development of new drug therapies to improve pregnancy outcome. On March 5-6, 2015, the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health sponsored a 2-day workshop titled Placental Origins of Adverse Pregnancy Outcomes: Potential Molecular Targets to begin to address this gap. Particular emphasis was given to the identification of important molecular pathways that could serve as drug targets and the advantages and disadvantages of targeting these particular pathways. This article is a summary of the proceedings of that workshop. A broad number of topics were covered that ranged from basic placental biology to clinical trials. This included research in the basic biology of placentation, such as trophoblast migration and spiral artery remodeling, and trophoblast sensing and response to infectious and noninfectious agents. Research findings in these areas will be critical for the formulation of the development of future treatments and the development of therapies for the prevention of a number of pregnancy disorders of placental origin that include preeclampsia, fetal growth restriction, and uterine inflammation. Research was also presented that summarized ongoing clinical efforts in the United States and in Europe that has tested novel interventions for preeclampsia and fetal growth restriction, including agents such as oral arginine supplementation, sildenafil, pravastatin, gene therapy with virally delivered vascular endothelial growth factor, and oxygen supplementation therapy. Strategies were also proposed to improve fetal growth by the enhancement of nutrient transport to the fetus by modulation of their placental transporters and the targeting of placental mitochondrial dysfunction and oxidative stress to improve placental health. The roles of microRNAs and placental-derived exosomes, as well as messenger RNAs, were also discussed in the context of their use for diagnostics and as drug targets. The workshop discussed the aspect of safety and pharmacokinetic profiles of potential existing and new therapeutics that will need to be determined, especially in the context of the unique pharmacokinetic properties of pregnancy and the hurdles and pitfalls of the translation of research findings into practice. The workshop also discussed novel methods of drug delivery and targeting during pregnancy with the use of macromolecular carriers, such as nanoparticles and biopolymers, to minimize placental drug transfer and hence fetal drug exposure. In closing, a major theme that developed from the workshop was that the scientific community must change their thinking of the pregnant woman and her fetus as a vulnerable patient population for which drug development should be avoided, but rather be thought of as a deprived population in need of more effective therapeutic interventions.
Collapse
Affiliation(s)
- John V Ilekis
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Ekaterini Tsilou
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Department of Health and Human Services, Bethesda, MD.
| | - Susan Fisher
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA
| | - Vikki M Abrahams
- Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine; New Haven, CT
| | - Michael J Soares
- Institute of Reproductive Health and Regenerative Medicine and Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - James C Cross
- Comparative Biology and Experimental Medicine, University of Calgary Health Sciences Centre, Calgary, Alberta, Canada
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Nicholas P Illsley
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack, NJ
| | - Leslie Myatt
- Center for Pregnancy and Newborn Research, University of Texas Health Science Center, San Antonio, TX
| | - Christine Colvis
- Therapeutics Discovery Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD
| | - Maged M Costantine
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - David M Haas
- Department of Obstetrics and Gynecology Indiana University, Indianapolis, IN
| | | | - Carl Weiner
- University of Kansas Medical Center, Kansas City, KS
| | - Erik Rytting
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX
| | - Gene Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
200
|
Beigi RH, Noguchi L, Brown G, Piper J, Watts DH. Performing Drug Safety Research During Pregnancy and Lactation: Biomedical HIV Prevention Research as a Template. J Womens Health (Larchmt) 2016; 25:761-6. [PMID: 23808668 PMCID: PMC4939378 DOI: 10.1089/jwh.2013.4398] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Evidence-based guidance regarding use of nearly all pharmaceuticals by pregnant and lactating women is limited. Models for performing research may assist in filling these knowledge gaps. Internationally, reproductive age women are at high risk of human immunodeficiency virus (HIV) acquisition. Susceptibility to HIV infection may be increased during pregnancy, and risk of maternal-child transmission is increased with incident HIV infection during pregnancy and lactation. A multidisciplinary meeting of experts was convened at the United States National Institutes of Health to consider paradigms for drug research in pregnancy and lactation applicable to HIV prevention. This report summarizes the meeting proceedings and describes a framework for research on candidate HIV prevention agent use during pregnancy and lactation that may also have broader applications to other pharmaceutical products.
Collapse
Affiliation(s)
- Richard H. Beigi
- Division of Reproductive Infectious Diseases, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Microbicide Trials Network, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Lisa Noguchi
- Microbicide Trials Network, Magee-Womens Research Institute, Pittsburgh, Pennsylvania
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Gina Brown
- Office of AIDS Research, National Institutes of Health, Bethesda, Maryland
| | - Jeanna Piper
- National Institute of Allergy and Infectious Diseases, Division of AIDS, National Institutes of Health, Bethesda, Maryland
| | - D. Heather Watts
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|