101
|
Schalkwijk S, Ter Heine R, Colbers A, Capparelli E, Best BM, Cressey TR, Greupink R, Russel FGM, Moltó J, Mirochnick M, Karlsson MO, Burger DM. Evaluating darunavir/ritonavir dosing regimens for HIV-positive pregnant women using semi-mechanistic pharmacokinetic modelling. J Antimicrob Chemother 2020; 74:1348-1356. [PMID: 30715324 DOI: 10.1093/jac/dky567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Darunavir 800 mg once (q24h) or 600 mg twice (q12h) daily combined with low-dose ritonavir is used to treat HIV-positive pregnant women. Decreased total darunavir exposure (17%-50%) has been reported during pregnancy, but limited data on unbound exposure are available. OBJECTIVES To evaluate total and unbound darunavir exposures following standard darunavir/ritonavir dosing and to explore the value of potential optimized darunavir/ritonavir dosing regimens for HIV-positive pregnant women. PATIENTS AND METHODS A population pharmacokinetic analysis was conducted based on data from 85 women. The final model was used to simulate total and unbound darunavir AUC0-τ and Ctrough during the third trimester of pregnancy, as well as to assess the probability of therapeutic exposure. RESULTS Simulations predicted that total darunavir exposure (AUC0-τ) was 24% and 23% lower in pregnancy for standard q24h and q12h dosing, respectively. Unbound darunavir AUC0-τ was 5% and 8% lower compared with post-partum for standard q24h and q12h dosing, respectively. The probability of therapeutic exposure (unbound) during pregnancy was higher for standard q12h dosing (99%) than for q24h dosing (94%). CONCLUSIONS The standard q12h regimen resulted in maximal and higher rates of therapeutic exposure compared with standard q24h dosing. Darunavir/ritonavir 600/100 mg q12h should therefore be the preferred regimen during pregnancy unless (adherence) issues dictate q24h dosing. The value of alternative dosing regimens seems limited.
Collapse
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
| | - Rob Ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud university medical center, Nijmegen, The Netherlands
| | - Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud university medical center, Nijmegen, The Netherlands
| | - Edmund Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences & School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Brookie M Best
- Skaggs School of Pharmacy and Pharmaceutical Sciences & School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Tim R Cressey
- Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Rick Greupink
- Department of Pharmacology & Toxicology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology & Toxicology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - José Moltó
- Fundació Lluita contra la Sida, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Mark Mirochnick
- Department of Pediatrics, Boston University School of Medicine, Boston, MA, USA
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - David M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences (RIHS), Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
102
|
Abduljalil K, Badhan RKS. Drug dosing during pregnancy-opportunities for physiologically based pharmacokinetic models. J Pharmacokinet Pharmacodyn 2020; 47:319-340. [PMID: 32592111 DOI: 10.1007/s10928-020-09698-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/20/2020] [Indexed: 12/15/2022]
Abstract
Drugs can have harmful effects on the embryo or the fetus at any point during pregnancy. Not all the damaging effects of intrauterine exposure to drugs are obvious at birth, some may only manifest later in life. Thus, drugs should be prescribed in pregnancy only if the expected benefit to the mother is thought to be greater than the risk to the fetus. Dosing of drugs during pregnancy is often empirically determined and based upon evidence from studies of non-pregnant subjects, which may lead to suboptimal dosing, particularly during the third trimester. This review collates examples of drugs with known recommendations for dose adjustment during pregnancy, in addition to providing an example of the potential use of PBPK models in dose adjustment recommendation during pregnancy within the context of drug-drug interactions. For many drugs, such as antidepressants and antiretroviral drugs, dose adjustment has been recommended based on pharmacokinetic studies demonstrating a reduction in drug concentrations. However, there is relatively limited (and sometimes inconsistent) information regarding the clinical impact of these pharmacokinetic changes during pregnancy and the effect of subsequent dose adjustments. Examples of using pregnancy PBPK models to predict feto-maternal drug exposures and their applications to facilitate and guide dose assessment throughout gestation are discussed.
Collapse
Affiliation(s)
- Khaled Abduljalil
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ, UK.
| | | |
Collapse
|
103
|
Murtagh R, Else LJ, Kuan KB, Khoo SH, Jackson V, Patel A, Lawler M, McDonald G, Le Blanc D, Avramovic G, Redmond N, Lambert JS. Therapeutic drug monitoring of darunavir/ritonavir in pregnancy. Antivir Ther 2020; 24:229-233. [PMID: 30728322 DOI: 10.3851/imp3291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Physiological changes during pregnancy can have a significant impact on antiretroviral pharmacokinetics (PK), which may result in reduced drug efficacy. Here we describe the PK of darunavir/ritonavir (DRV/r) 800/100 once daily in a cohort of pregnant women undergoing routine therapeutic drug monitoring (TDM) as well as transplacental passage of DRV by measuring and comparing cord blood and maternal blood samples at delivery. METHODS Pregnant HIV-positive women received DRV/r as part of routine pre-natal care. Demographic and clinical data were collected. DRV plasma concentrations [DRV] were determined in the first (T1), second (T2) and third (T3) trimester and at postpartum (PP). The target concentration was 550 ng/ml. Where possible, paired maternal and cord blood samples were taken at delivery. RESULTS A total of 33 women were enrolled. Samples were taken 14-20 h post-dose and measured concentrations were extrapolated to 24 h post-dose. At the time nearest to delivery, all but four had undetectable plasma viral loads (pVL). [DRV] were determined in 1 (T1); 14 (T2); 32 (T3) and 29 (PP). 1 sample was <550 ng/ml at T2, 6 at T3 and 3 at PP. [DRV] were significantly lower at T2/T3 relative to PP. CONCLUSIONS [DRV] in T2 and T3 were 36-55% when compared with PP. However, DRV PK in pregnancy were not associated with a lack of virological suppression at delivery as of the 33 patients enrolled in this study, 31 had no HIV transmission from mother to child. Data regarding two candidates were not available as they delivered in a separate health-care facility.
Collapse
Affiliation(s)
- Ross Murtagh
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Laura J Else
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Kenneth Bk Kuan
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Saye H Khoo
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | | | - Anjali Patel
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | | | | | | | - Gordana Avramovic
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.,Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Niamh Redmond
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - John S Lambert
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland.,Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland.,The Rotunda Hospital, Dublin, Ireland
| |
Collapse
|
104
|
Puhto T, Kokki M, Hakomäki H, Spalding M, Gunnar T, Alahuhta S, Vakkala M. Single dose epidural hydromorphone in labour pain: maternal pharmacokinetics and neonatal exposure. Eur J Clin Pharmacol 2020; 76:969-977. [PMID: 32363420 PMCID: PMC7306027 DOI: 10.1007/s00228-020-02880-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/24/2020] [Indexed: 11/02/2022]
Abstract
INTRODUCTION Epidural hydromorphone could be useful in obstetric analgesia as there is a need for a more water-soluble opioid than sufentanil or fentanyl with prolonged analgesic effect. To our knowledge, the pharmacokinetics of epidural hydromorphone has not been evaluated in parturients. MATERIALS AND METHODS In this pilot study, seven healthy parturients were given a single epidural dose of hydromorphone for labour pain. One parturient received 1.5 mg, two 0.75 mg and four 0.5 mg of hydromorphone hydrochloride. Dose was decreased due to nausea and pruritus. Hydromorphone's effect, adverse effects and plasma concentrations were evaluated. Neonatal drug exposure was evaluated by umbilical vein and artery opioid concentration at birth. Neonatal outcomes were assessed using Apgar and the Neurologic Adaptive Capacity Score (NACS). RESULTS All patients received additional levobupivacaine doses on parturients' requests. The first dose was requested at a median of 163 min (range 19-303 min) after hydromorphone administration. A total of 12 opioid related expected adverse events were reported by seven parturients. All newborn outcomes were uneventful. Hydromorphone's distribution and elimination after single epidural dose seem similar to that reported for non-pregnant subjects after intravenous hydromorphone administration, but further research is required to confirm this observation. CONCLUSIONS The optimal dose of hydromorphone in labour pain warrants further evaluation.
Collapse
Affiliation(s)
- Terhi Puhto
- Department of Anaesthesiology, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital, PO Box 21, 90029, Oulu, Finland.
| | - Merja Kokki
- Department of Anaesthesiology and Intensive Care, Kuopio University Hospital, Kuopio, Finland
| | | | - Michael Spalding
- Department of Anaesthesiology, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital, PO Box 21, 90029, Oulu, Finland
| | - Teemu Gunnar
- Forensic Toxicology Unit (THL), The Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Seppo Alahuhta
- Department of Anaesthesiology, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital, PO Box 21, 90029, Oulu, Finland
| | - Merja Vakkala
- Department of Anaesthesiology, Medical Research Center Oulu (MRC Oulu), Oulu University Hospital, PO Box 21, 90029, Oulu, Finland
| |
Collapse
|
105
|
Neary M, Owen A, Olagunju A. Pharmacokinetics of HIV therapies in pregnant patients: an update. Expert Opin Drug Metab Toxicol 2020; 16:449-461. [PMID: 32271621 DOI: 10.1080/17425255.2020.1754792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Mother-to-child transmission (MTCT) of HIV is thought to account for over 90% of new pediatric infections, and is associated with poor maternal and fetal outcomes. As such ensuring further reduction in MTCT is a priority in HIV treatment and prevention programs. AREAS COVERED This review aims to provide a comprehensive update on the pharmacokinetics of recently approved antiretroviral drugs and novel drug formulations and delivery systems. Alongside recent recommendations for dose adjustments, and an overview of the implications of co-infections on the pharmacokinetics of antiretrovirals relevant to pregnant HIV positive patients. Additionally, potential opportunities to progress pharmacokinetic research of new treatments in this population are highlighted. EXPERT OPINION In order to improve our understanding of how to provide safe and effective treatment to HIV positive pregnant women, further work is required to enable their inclusion in early stages of clinical trials. Incentives must be created for this research, in the form of additional investment by key stakeholders and regulatory agencies. Furthermore, as the incidence of MTCT is reduced globally there is a need to conduct long-term pharmacovigilance studies in uninfected children exposed to HIV and antiretrovirals in utero, in order to determine the safest and most effective antiretroviral therapies.
Collapse
Affiliation(s)
- Megan Neary
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool , Liverpool, UK
| | - Andrew Owen
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool , Liverpool, UK
| | - Adeniyi Olagunju
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool , Liverpool, UK.,Faculty of Pharmacy, Obafemi Awolowo University , Ile-Ife, Nigeria
| |
Collapse
|
106
|
Abstract
Pharmacologic interventions play a major role in obstetrical care throughout pregnancy, labor and delivery and the postpartum. Traditionally, obstetrical providers have utilized standard dosing regimens developed for non-obstetrical indications based on pharmacokinetic knowledge from studies in men or non-pregnant women. With the recognition of pregnancy as a special pharmacokinetic population in the late 1990s, investigators have begun to study drug disposition in this unique patient dyad. Many of the basic physiologic changes that occur during pregnancy have significant impact on drug absorption, distribution and clearance. Activity of Phase I and Phase II drug metabolizing enzymes are differentially altered by pregnancy, resulting in drug concentrations sufficiently different for some medications that efficacy or toxicity is affected. Placental transporters play a major dynamic role in determining fetal drug exposure. In the past two decades, we have begun to expand our understanding of obstetrical pharmacology; however, to truly optimize pharmacologic care of our pregnant patients and their developing fetus, additional research is critically needed.
Collapse
|
107
|
Abstract
Hepatic drug metabolism is a major route of drug elimination, mediated by multiple drug-metabolizing enzymes. Any changes in the rate and extent of hepatic drug metabolism can lead to altered drug efficacy or toxicity. Accumulating clinical evidence indicates that pregnancy is accompanied by changes in hepatic drug metabolism. In this article, we discuss in vitro and in vivo tools used to study the mechanisms underlying the altered drug metabolism during pregnancy, focusing on primary hepatocyte culture, transgenic animal models, and use of probe drugs to assess change in enzymatic activity. The information obtained from these studies has enabled prediction of clinical PK changes for a given drug in pregnant women.
Collapse
Affiliation(s)
- Hyunyoung Jeong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, United States.
| | - Catherine S. Stika
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
108
|
Liao MZ, Flood Nichols SK, Ahmed M, Clark S, Hankins GD, Caritis S, Venkataramanan R, Haas D, Quinney SK, Haneline LS, Tita AT, Manuck T, Wang J, Thummel KE, Brown LM, Ren Z, Easterling TR, Hebert MF. Effects of Pregnancy on the Pharmacokinetics of Metformin. Drug Metab Dispos 2020; 48:264-271. [PMID: 31980499 PMCID: PMC7076518 DOI: 10.1124/dmd.119.088435] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/30/2019] [Indexed: 12/22/2022] Open
Abstract
This study's primary objective was to fully characterize the pharmacokinetics of metformin in pregnant women with gestational diabetes mellitus (GDM) versus nonpregnant controls. Steady-state oral metformin pharmacokinetics in pregnant women with GDM receiving either metformin monotherapy (n = 24) or a combination with glyburide (n = 30) as well as in nonpregnant women with type 2 diabetes mellitus (T2DM) (n = 24) were determined utilizing noncompartmental techniques. Maternal and umbilical cord blood samples were collected at delivery from 38 women. With both 500- and 1000-mg doses, metformin bioavailability, volume of distribution beta (V β ), clearance, and renal clearance were significantly increased during pregnancy. In addition, in the women receiving metformin 500 mg, significantly higher metformin apparent oral clearance (CL/F) (27%), weight-adjusted renal secretion clearance (64%), and apparent oral volume of distribution beta (V β /F) (33%) were seen during pregnancy. Creatinine clearance was significantly higher during pregnancy. Increasing metformin dose from 500 to 1000 mg orally twice daily significantly increased V β /F by 28%, weight-adjusted V β /F by 32% and CL/F by 25%, and weight-adjusted CL/F by 28% during pregnancy. Mean metformin umbilical cord arterial-to-venous plasma concentration ratio was 1.0 ± 0.1, venous umbilical cord-to-maternal concentration ratio was 1.4 ± 0.5, and arterial umbilical cord-to-maternal concentration ratio was 1.5 ± 0.5. Systemic exposure after a 500-mg dose of metformin was lower during pregnancy compared with the nonpregnant women with T2DM. However, in patients receiving metformin 1000 mg, changes in estimated bioavailability during pregnancy offset the changes in clearance leading to no significant change in CL/F with the higher dose. SIGNIFICANCE STATEMENT: Gestational diabetes mellitus complicates 5%-13% of pregnancies and is often treated with metformin. Pregnant women undergo physiological changes that alter drug disposition. Preliminary data suggest that pregnancy lowers metformin concentrations, potentially affecting efficacy and safety. This study definitively describes pregnancy's effects on metformin pharmacokinetics and expands the mechanistic understanding of pharmacokinetic changes across the dosage range. Here we report the nonlinearity of metformin pharmacokinetics and the increase in bioavailability, clearance, renal clearance, and volume of distribution during pregnancy.
Collapse
Affiliation(s)
- Michael Z Liao
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Shannon K Flood Nichols
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Mahmoud Ahmed
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Shannon Clark
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Gary D Hankins
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Steve Caritis
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Raman Venkataramanan
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - David Haas
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Sara K Quinney
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Laura S Haneline
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Alan T Tita
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Tracy Manuck
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Joanne Wang
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Kenneth E Thummel
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Linda Morris Brown
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Zhaoxia Ren
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Thomas R Easterling
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Mary F Hebert
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| |
Collapse
|
109
|
Abstract
Pregnant women frequently take prescription and over the counter medications. The efficacy of medications is affected by the many physiological changes during pregnancy, and these events may be further impacted by genetic factors. Research on pharmacogenomic and pharmacokinetic influences on drug disposition during pregnancy has lagged behind other fields. Clinical investigators have demonstrated altered activity of several drug metabolizing enzymes during pregnancy. Emerging evidence also supports the influence of pharmacogenomic variability in drug response for many important classes of drugs commonly used in pregnancy. Prescribing medications during pregnancy requires an understanding of the substantial dynamic physiologic and metabolic changes that occur during gestation. Pharmacogenomics also contributes to the inter-individual variability in response to many medications, and more research is needed to understand how best to manage drug therapy in pregnant women.
Collapse
Affiliation(s)
- Hannah K Betcher
- Department of Psychiatry, Northwestern University Feinberg School of Medicine, 676N St. Clair St. Ste 1000, Chicago IL, USA; Mayo Clinic, Rochester, MN, USA.
| | - Alfred L George
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Searle 8-510, 320 East Superior Street, Chicago, IL 60611, USA.
| |
Collapse
|
110
|
Hagen M, Alchin J. Nonprescription drugs recommended in guidelines for common pain conditions. Pain Manag 2020; 10:117-129. [DOI: 10.2217/pmt-2019-0057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Evidence-based pain guidelines allow recommendation of nonprescription analgesics to patients, facilitating self-care. We researched clinical practice guidelines for common conditions on websites of pain associations, societies, health institutions and organizations, PubMed, ProQuest, Embase, Google Scholar until April 2019. We wanted to determine whether there is a consensus between guidelines. From 114 identified guidelines, migraine (27) and osteoarthritis (26) have been published most around the world, while dysmenorrhea (14) is mainly discussed in developing countries. Specific recommendations to pregnant women, children and older people predominantly come from the UK and USA. We found that acetaminophen and oral nonsteroidal anti-inflammatory drugs (NSAIDs) represent first-line management across all pain conditions in adults and children. In osteoarthritis, topical NSAIDs should be considered before oral NSAIDs. This knowledge might persuade patients that using these drugs first could enable fast and effective pain relief.
Collapse
Affiliation(s)
| | - John Alchin
- Pain Management Centre, Burwood Hospital, Christchurch, New Zealand
| |
Collapse
|
111
|
Population Pharmacokinetics of Isoniazid, Pyrazinamide, and Ethambutol in Pregnant South African Women with Tuberculosis and HIV. Antimicrob Agents Chemother 2020; 64:AAC.01978-19. [PMID: 31844002 DOI: 10.1128/aac.01978-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/05/2019] [Indexed: 11/20/2022] Open
Abstract
Tuberculosis is an important cause of maternal morbidity, but little is known about the effects of pregnancy on antituberculosis drug concentrations. We developed population pharmacokinetic models to describe drug dispositions of isoniazid, pyrazinamide, and ethambutol in pregnant women with tuberculosis and HIV. HIV-positive pregnant women with tuberculosis receiving standard first-line tuberculosis treatment and participating in Tshepiso, a prospective cohort study in Soweto, South Africa, underwent sparse pharmacokinetic sampling at >36 weeks of gestation and 7 weeks postpartum. The effects of pregnancy on the pharmacokinetics of isoniazid, pyrazinamide, and ethambutol were investigated via population pharmacokinetic modeling. Isoniazid, pyrazinamide, and ethambutol concentrations were available for 29, 18, and 18 women, respectively. Their median weight was 66 kg while pregnant and 64 kg postpartum. No significant differences were observed in drug clearance, volume of distribution, or bioavailability during and after pregnancy. The model-estimated isoniazid, pyrazinamide, and ethambutol area under the concentration-time curve from 0 to 24 h (AUC0-24) medians were, respectively, 6.88, 419, and 16.5 mg · h/liter during pregnancy versus 5.01, 407, and 19.0 mg · h/liter postpartum. The model-estimated maximum concentration (C max) medians for isoniazid, pyrazinamide, and ethambutol were, respectively, 1.39, 35.9, and 1.82 mg/liter during pregnancy versus 1.43, 34.5, and 2.11 mg/liter postpartum. A posteriori power calculations determined that our analysis was powered 91.8%, 59.2%, and 90.1% at a P of <0.01 to detect a 40% decrease in the AUCs of isoniazid, pyrazinamide, and ethambutol, respectively. Pregnancy does not appear to cause relevant changes in the exposure to isoniazid, pyrazinamide, and ethambutol. Additional studies of antituberculosis drugs in pregnancy are needed.
Collapse
|
112
|
A Randomized Controlled Trial of Three- versus Five-Day Artemether-Lumefantrine Regimens for Treatment of Uncomplicated Plasmodium falciparum Malaria in Pregnancy in Africa. Antimicrob Agents Chemother 2020; 64:AAC.01140-19. [PMID: 31818818 PMCID: PMC7038309 DOI: 10.1128/aac.01140-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/24/2019] [Indexed: 01/09/2023] Open
Abstract
Artemether-lumefantrine antimalarial efficacy in pregnancy could be compromised by reduced drug exposure. Population-based simulations suggested that therapeutic efficacy would be improved if the treatment duration was increased. Artemether-lumefantrine antimalarial efficacy in pregnancy could be compromised by reduced drug exposure. Population-based simulations suggested that therapeutic efficacy would be improved if the treatment duration was increased. We assessed the efficacy, tolerability, and pharmacokinetics of an extended 5-day regimen of artemether-lumefantrine compared to the standard 3-day treatment in 48 pregnant women and 48 nonpregnant women with uncomplicated falciparum malaria in an open-label, randomized clinical trial. Babies were assessed at birth and 1, 3, 6, and 12 months. Nonlinear mixed-effects modeling was used to characterize the plasma concentration-time profiles of artemether and lumefantrine and their metabolites. Both regimens were highly efficacious (100% PCR-corrected cure rates) and well tolerated. Babies followed up to 1 year had normal development. Parasite clearance half-lives were longer in pregnant women (median [range], 3.30 h [1.39 to 7.83 h]) than in nonpregnant women (2.43 h [1.05 to 6.00 h]) (P=0.005). Pregnant women had lower exposures to artemether and dihydroartemisinin than nonpregnant women, resulting in 1.2% decreased exposure for each additional week of gestational age. By term, these exposures were reduced by 48% compared to nonpregnant patients. The overall exposure to lumefantrine was improved with the extended regimen, with no significant differences in exposures to lumefantrine or desbutyl-lumefantrine between pregnant and nonpregnant women. The extended artemether-lumefantrine regimen was well tolerated and safe and increased the overall antimalarial drug exposure and so could be a promising treatment option in pregnancy in areas with lower rates of malaria transmission and/or emerging drug resistance. (This study has been registered at ClinicalTrials.gov under identifier NCT01916954.)
Collapse
|
113
|
Population Pharmacokinetics of Sulindac and Genetic Polymorphisms of FMO3 and AOX1 in Women with Preterm Labor. Pharm Res 2020; 37:44. [PMID: 31993760 DOI: 10.1007/s11095-020-2765-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 01/16/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE This prospective study aimed to evaluate the effects of genetic polymorphisms in sulindac-related metabolizing enzyme genes including FMO3 and AOX1 on the population pharmacokinetics of sulindac in 58 pregnant women with preterm labor. METHODS Plasma samples were collected at 1.5, 4, and 10 h after first oral administration of sulindac. Plasma concentrations of sulindac and its active metabolite (sulindac sulfide) were determined, and pharmacokinetic analysis was performed with NONMEM 7.3. RESULTS The mean maternal and gestational ages at the time of dosing were 32.5 ± 4.4 (range, 20-41) years and 27.4 ± 4.4 (range, 16.4-33.4) weeks, respectively. In the population pharmacokinetic analysis, one depot compartment model of sulindac with absorption lag time best described the data. The metabolism of sulindac and sulindac sulfide was described using Michaelis-Menten kinetics. In stepwise modeling, gestational age impacted volume of distribution (Vc), and FMO3 rs2266782 was shown by the Michaelis constant to affect conversion of sulindac sulfide to sulindac (KM32); these were retained in the final model. CONCLUSIONS Genetic polymorphisms of FMO3 and AOX1 could affect the pharmacokinetics of sulindac in women who undergo preterm labor. The results of this study could help clinicians develop individualized treatment plans for administering sulindac.
Collapse
|
114
|
Loukotková L, Basavarajappa M, Lumen A, Roberts R, Mattison D, Morris SM, Fisher J, Beland FA, Gamboa da Costa G. Pharmacokinetics of oseltamivir phosphate and oseltamivir carboxylate in non-pregnant and pregnant rhesus monkeys. Regul Toxicol Pharmacol 2020; 112:104569. [PMID: 31927005 DOI: 10.1016/j.yrtph.2019.104569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/24/2019] [Indexed: 11/25/2022]
Abstract
Oseltamivir is an antiviral drug approved to treat influenza in humans. Although the dosing regimen of this drug is well established for non-pregnant patients, it is not clear if the significant physiological alterations associated with pregnancy affect the pharmacokinetics of oseltamivir and, thus, warrant different dosing regimens to assure efficacy. In this study, we investigated the suitability of rhesus macaques as an animal model for studying oseltamivir pharmacokinetics during all trimesters of pregnancy in comparison to pre-pregnant conditions. Specifically, we compared the pharmacokinetics of oseltamivir and its pharmacologically active metabolite oseltamivir carboxylate in rhesus monkeys after intravenous and nasogastric administration of 2.5 mg oseltamivir phosphate/kg body weight given prior to and during the first, second, and third trimesters of pregnancy. Pregnancy had only a modest effect upon the pharmacokinetic parameters of oseltamivir and oseltamivir carboxylate. Monkeys treated intravenously in the third trimester had a reduction in Vd and CL, compared to non-pregnant monkeys. These changes did not occur in the other two trimesters. Pregnant monkeys treated intravenously had 20-25% decrease in AUC0-∞ of oseltamivir carboxylate and a corresponding increase in Vd and CL. Pregnant monkeys treated nasogastrically with oseltamivir phosphate demonstrated a pattern that recapitulated intravenous dosing. Taken together these data indicate that rhesus monkeys are an acceptable model for studying drug-pregnancy interactions.
Collapse
Affiliation(s)
- Lucie Loukotková
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | | | - Annie Lumen
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | - Rosemary Roberts
- FDA Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - Donald Mattison
- Risk Sciences International, Ottawa, Ontario, Canada and Hilton Head Island, SC USA and University of Ottawa, Ontario, Canada
| | - Suzanne M Morris
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | - Jeffrey Fisher
- FDA National Center for Toxicological Research, Jefferson, AR, USA
| | | | | |
Collapse
|
115
|
Betcher HK, Wisner KL. Psychotropic Treatment During Pregnancy: Research Synthesis and Clinical Care Principles. J Womens Health (Larchmt) 2019; 29:310-318. [PMID: 31800350 DOI: 10.1089/jwh.2019.7781] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Psychiatric illnesses are common in women of childbearing age. The perinatal period is a particularly high-risk time for depression, bipolar, and anxiety disorders. Methods: The scope of the public health problem of perinatal mental disorders is discussed followed by an examination of the specific research methods utilized for the study of birth and developmental outcomes associated with maternal mental illness and its treatment. The evidence on exposure to common psychotropics during pregnancy and breastfeeding is reviewed. Results: Selective serotonin reuptake inhibitors or serotonin-norepinephrine reuptake inhibitor medications are not associated with higher rates of birth defects or long-term changes in mental development after adjustment for confounding factors associated with underlying psychiatric illness. Lithium exposure is associated with an increased risk for fetal cardiac malformations, but this risk is lower than previously thought (absolute risk of Ebstein's anomaly 6/1,000). Antipsychotics, other than risperidone and potentially paliperidone, have not been associated with an increase in birth defects; olanzapine and quetiapine have been linked with an elevated risk of gestational diabetes. Due to the dramatic physiological changes of pregnancy and enhanced hepatic metabolism, drug doses may need to be adjusted during pregnancy to sustain efficacy. Untreated maternal psychiatric illness also carries substantial risks for the mother, fetus, infant, and family. Conclusions: The goal of perinatal mental health treatment is to optimally provide pharmacotherapy to mitigate the somatic and psychosocial burdens of maternal psychiatric disorders. Regular symptom monitoring during pregnancy and postpartum and medication dose adjustments to sustain efficacy constitutes good practice.
Collapse
Affiliation(s)
- Hannah K Betcher
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
| | - Katherine L Wisner
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
116
|
Mutagonda RF, Minzi OMS, Massawe SN, Asghar M, Färnert A, Kamuhabwa AAR, Aklillu E. Pregnancy and CYP3A5 Genotype Affect Day 7 Plasma Lumefantrine Concentrations. Drug Metab Dispos 2019; 47:1415-1424. [PMID: 31744845 DOI: 10.1124/dmd.119.088062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/27/2019] [Indexed: 12/21/2022] Open
Abstract
Pregnancy and pharmacogenetics variation alter drug disposition and treatment outcome. The objective of this study was to investigate the effect of pregnancy and pharmacogenetics variation on day 7 lumefantrine (LF) plasma concentration and therapeutic responses in malaria-infected women treated with artemether-lumefantrine (ALu) in Tanzania. A total of 277 (205 pregnant and 72 nonpregnant) women with uncomplicated Plasmodium falciparum malaria were enrolled. Patients were treated with ALu and followed up for 28 days. CYP3A4, CYP3A5, and ABCB1 genotyping were done. Day 7 plasma LF concentration and the polymerase chain reaction (PCR) - corrected adequate clinical and parasitological response (ACPR) at day 28 were determined. The mean day 7 plasma LF concentrations were significantly lower in pregnant women than nonpregnant women [geometric mean ratio = 1.40; 95% confidence interval (CI) of geometric mean ratio (1.119-1.1745), P < 0.003]. Pregnancy, low body weight, and CYP3A5*1/*1 genotype were significantly associated with low day 7 LF plasma concentration (P < 0.01). PCR-corrected ACPR was 93% (95% CI = 89.4-96.6) in pregnant women and 95.7% (95% CI = 90.7-100) in nonpregnant women. Patients with lower day 7 LF concentration had a high risk of treatment failure (mean 652 vs. 232 ng/ml, P < 0.001). In conclusion, pregnancy, low body weight, and CYP3A5*1 allele are significant predictors of low day 7 LF plasma exposure. In turn, lower day 7 LF concentration is associated with a higher risk of recrudescence. SIGNIFICANCE STATEMENT: This study reports a number of factors contributing to the lower day 7 lumefantrine (LF) concentration in women, which includes pregnancy, body weight, and CYP3A5*1/*1 genotype. It also shows that day 7 LF concentration is a main predictor of malaria treatment. These findings highlight the need to look into artemether-LF dosage adjustment in pregnant women so as to be able to maintain adequate drug concentration, which is required to reduce treatment failure rates in pregnant women.
Collapse
MESH Headings
- Antimalarials/administration & dosage
- Antimalarials/blood
- Antimalarials/therapeutic use
- Artemether, Lumefantrine Drug Combination/administration & dosage
- Artemether, Lumefantrine Drug Combination/blood
- Artemether, Lumefantrine Drug Combination/therapeutic use
- Cohort Studies
- Cytochrome P-450 CYP3A/genetics
- Dose-Response Relationship, Drug
- Female
- Gene Frequency
- Genotype
- Humans
- Malaria, Falciparum/blood
- Malaria, Falciparum/drug therapy
- Malaria, Falciparum/genetics
- Pregnancy
- Pregnancy Complications, Parasitic/blood
- Pregnancy Complications, Parasitic/drug therapy
- Pregnancy Complications, Parasitic/genetics
- Pregnancy Trimester, Third
- Prospective Studies
- Time Factors
Collapse
Affiliation(s)
- Ritah F Mutagonda
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Omary M S Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Siriel N Massawe
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Muhammad Asghar
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Anna Färnert
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Appolinary A R Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| | - Eleni Aklillu
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy (R.F.M., O.O.M.S.M., A.A.R.K.), and Department of Obstetrics and Gynecology, School of Medicine (S.N.M.), Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania; Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (M.A., A.F.); Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden (A.F.); and Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Stockholm, Sweden (E.A.)
| |
Collapse
|
117
|
Abstract
OPINION STATEMENT Despite the increase of breast cancer incidence with age, approximately 7 to 10% ofwomen diagnosed with breast cancer are younger than the age 40. This subgroup ofpatients has different risk factors, tumour biology, clinical outcomes, and specific psy- chosocial issues, such as fertility preservation, family planning, and job reintegration. However, age alone should not be the main consideration when choosing the aggressive- ness of the treatment, as other factors must be considered, including the biologic aggressiveness of the tumour, potential long-term toxicities, and the preferences of the patient. Fertility preservation techniques should be discussed with the patient before starting any cancer treatment. Despite the significant percentage of breast cancer patients younger than age 40, fewclinical studies have specifically investigated disease characteristics and outcomes of this population, and most therapies routinely administered to these younger women were tested in older patients. Moreover, young women who have breast cancer are at a greater risk of sexual and psychological distress, and clinicians should address these issues in order to properly support patients during the long diagnostic and therapeutic journey. Consequently, it is essential to follow diagnostic and treatment guidelines specificallyaddressed to young women. Additional specific procedures should be followed to treat pregnant patients with breast cancer.
Collapse
|
118
|
Souza MCO, Marques MP, Duarte G, Lanchote VL. Pharmacokinetics and Placental Transfer of Bupivacaine Enantiomers in HIV-Infected Parturient Women on Antiretroviral Therapy. J Clin Pharmacol 2019; 60:566-572. [PMID: 31696528 DOI: 10.1002/jcph.1554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 10/17/2019] [Indexed: 11/05/2022]
Abstract
Bupivacaine, a local anesthetic, is commercialized as a racemic mixture of R-bupivacaine and S-bupivacaine enantiomers. HIV infection increases the expression of placental P-glycoprotein (P-gp), and antiretroviral (ARV) therapy inhibits cytochrome P450 3A and P-gp. The present study evaluates the kinetic disposition of bupivacaine enantiomers in HIV-infected parturient women on ARV therapy. In this study, HIV-infected parturient women (n = 10) treated with zidovudine, lamivudine, lopinavir, and ritonavir were investigated. Anesthesia and/or analgesia was achieved by the administration of 0.5% racemic bupivacaine hydrochloride with 1:200000 epinephrine in the epidural space at doses of 2.5 to 22.5 mg. Maternal serial blood samples were obtained at the time immediately before and 5, 15, 30, 45, and 60 minutes and 2, 4, 6, 8, 10, 12, and 14 hours after administration of the bupivacaine. At the time of delivery, samples of maternal and umbilical cord vein blood were also collected. The results suggest that bupivacaine pharmacokinetics are enantioselective, revealing higher maternal plasma concentrations of the R-bupivacaine enantiomer (area under the total plasma concentration-time curve was calculated by the trapezoid method with extrapolation to infinity/dose(S)/(R) = 0.91; P < .05). The plasma unbound fraction of the drug (0.09 vs 0.06) and the umbilical cord vein/maternal plasma ratio (0.47 vs 0.39) were higher for the R-bupivacaine enantiomer than the S-bupivacaine enantiomer (P < .05). ARV therapy with ritonavir confers an enantioselective interaction between the enantiomers of bupivacaine and placental P-gp, producing greater inhibition of efflux transport of the R-bupivacaine enantiomer. Possible changes in the well-being of the fetuses of mothers under analgesia may be a consequence of the increased placental transfer of bupivacaine enantiomers to the fetal circulation.
Collapse
Affiliation(s)
- Marília Cristina Oliveira Souza
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Maria Paula Marques
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Geraldo Duarte
- Department of Gynecology and Obstetrics, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| | - Vera Lucia Lanchote
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil
| |
Collapse
|
119
|
Gupta A, Montepiedra G, Aaron L, Theron G, McCarthy K, Bradford S, Chipato T, Vhembo T, Stranix-Chibanda L, Onyango-Makumbi C, Masheto GR, Violari A, Mmbaga BT, Aurpibul L, Bhosale R, Mave V, Rouzier V, Hesseling A, Shin K, Zimmer B, Costello D, Sterling TR, Chakhtoura N, Jean-Philippe P, Weinberg A. Isoniazid Preventive Therapy in HIV-Infected Pregnant and Postpartum Women. N Engl J Med 2019; 381:1333-1346. [PMID: 31577875 PMCID: PMC7051859 DOI: 10.1056/nejmoa1813060] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The safety, efficacy, and appropriate timing of isoniazid therapy to prevent tuberculosis in pregnant women with human immunodeficiency virus (HIV) infection who are receiving antiretroviral therapy are unknown. METHODS In this multicenter, double-blind, placebo-controlled, noninferiority trial, we randomly assigned pregnant women with HIV infection to receive isoniazid preventive therapy for 28 weeks, initiated either during pregnancy (immediate group) or at week 12 after delivery (deferred group). Mothers and infants were followed through week 48 after delivery. The primary outcome was a composite of treatment-related maternal adverse events of grade 3 or higher or permanent discontinuation of the trial regimen because of toxic effects. The noninferiority margin was an upper boundary of the 95% confidence interval for the between-group difference in the rate of the primary outcome of less than 5 events per 100 person-years. RESULTS A total of 956 women were enrolled. A primary outcome event occurred in 72 of 477 women (15.1%) in the immediate group and in 73 of 479 (15.2%) in the deferred group (incidence rate, 15.03 and 14.93 events per 100 person-years, respectively; rate difference, 0.10; 95% confidence interval [CI], -4.77 to 4.98, which met the criterion for noninferiority). Two women in the immediate group and 4 women in the deferred group died (incidence rate, 0.40 and 0.78 per 100 person-years, respectively; rate difference, -0.39; 95% CI, -1.33 to 0.56); all deaths occurred during the postpartum period, and 4 were from liver failure (2 of the women who died from liver failure had received isoniazid [1 in each group]). Tuberculosis developed in 6 women (3 in each group); the incidence rate was 0.60 per 100 person-years in the immediate group and 0.59 per 100 person-years in the deferred group (rate difference, 0.01; 95% CI, -0.94 to 0.96). There was a higher incidence in the immediate group than in the deferred group of an event included in the composite adverse pregnancy outcome (stillbirth or spontaneous abortion, low birth weight in an infant, preterm delivery, or congenital anomalies in an infant) (23.6% vs. 17.0%; difference, 6.7 percentage points; 95% CI, 0.8 to 11.9). CONCLUSIONS The risks associated with initiation of isoniazid preventive therapy during pregnancy appeared to be greater than those associated with initiation of therapy during the postpartum period. (Funded by the National Institutes of Health; IMPAACT P1078 TB APPRISE ClinicalTrials.gov number, NCT01494038.).
Collapse
Affiliation(s)
- Amita Gupta
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Grace Montepiedra
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Lisa Aaron
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Gerhard Theron
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Katie McCarthy
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Sarah Bradford
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Tsungai Chipato
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Tichaona Vhembo
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Lynda Stranix-Chibanda
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Carolyne Onyango-Makumbi
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Gaerolwe R Masheto
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Avy Violari
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Blandina T Mmbaga
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Linda Aurpibul
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Ramesh Bhosale
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Vidya Mave
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Vanessa Rouzier
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Anneke Hesseling
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Katherine Shin
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Bonnie Zimmer
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Diane Costello
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Timothy R Sterling
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Nahida Chakhtoura
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Patrick Jean-Philippe
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| | - Adriana Weinberg
- From the Center for Clinical Global Health Education, Johns Hopkins University, Baltimore (A.G., V.M.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (K.S., P.J.-P.), and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (N.C.), National Institutes of Health, Bethesda - all in Maryland; the Harvard T.H. Chan School of Public Health, Boston (G.M., L. Aaron, G.R.M.); the Family Clinical Research Unit, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town (G.T.), the Perinatal HIV Research Unit, University of the Witwatersrand, Johannesburg (A.V.), and the Desmond Tutu TB Center, Department of Paediatrics and Child Health, Stellenbosch University, Tygerberg (A.H.) - all in South Africa; FHI 360, Durham, NC (K.M., S.B.); University of Zimbabwe College of Health Sciences Clinical Trials Research Centre, Harare (T.C., T.V., L.S.-C.); Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda (C.O.-M.); Botswana Harvard AIDS Institute Partnership, Gaborone (G.R.M.); Kilimanjaro Christian Medical Centre, Moshi, Tanzania (B.T.M.); Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand (L. Aurpibul); Byramjee Jeejeebhoy Government Medical College (R.B.) and Byramjee Jeejeebhoy Government College-Johns Hopkins Clinical Research Site (A.G., V.M.), Pune, India; Les Centres GHESKIO Clinical Research Site (GHESKIO-INLR), Port au Prince, Haiti (V.R.); Frontier Science Foundation, Amherst, NY (B.Z.); University of California, Los Angeles, Los Angeles (D.C.); Vanderbilt University Medical Center, Nashville (T.R.S.); and the University of Colorado Denver Anschutz Medical Campus, Aurora (A.W.)
| |
Collapse
|
120
|
Colbers A, Mirochnick M, Schalkwijk S, Penazzato M, Townsend C, Burger D. Importance of Prospective Studies in Pregnant and Breastfeeding Women Living With Human Immunodeficiency Virus. Clin Infect Dis 2019; 69:1254-1258. [PMID: 30783649 PMCID: PMC6743813 DOI: 10.1093/cid/ciz121] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/08/2019] [Indexed: 12/02/2022] Open
Abstract
Recently, the US Food and Drug Administration and European Medicines Agency issued warnings on the use of dolutegravir and darunavir/cobicistat for treatment of pregnant women living with human immunodeficiency virus (HIV). It took 3-5 years to identify the risks associated with the use of these antiretroviral drugs, during which time pregnant women were exposed to these drugs in clinical care, outside of controlled clinical trial settings. Across all antiretroviral drugs, the interval between registration of new drugs and first data on pharmacokinetics and safety in pregnancy becoming available is around 6 years. In this viewpoint, we provide considerations for clinical pharmacology research to provide safe and effective treatment of pregnant and breastfeeding women living with HIV and their children. These recommendations will lead to timelier availability of safety and pharmacokinetic information needed to develop safe treatment strategies for pregnant and breastfeeding women living with HIV, and are applicable to other chronic disease areas requiring medication during pregnancy.
Collapse
Affiliation(s)
- Angela Colbers
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Stein Schalkwijk
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Claire Townsend
- HIV Department, World Health Organization, Geneva, Switzerland
- Population, Policy and Practice Programme, UCL Great Ormond Street Institute of Child Health, University College London, United Kingdom
| | - David Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
121
|
Shanmugalingam R, Wang X, Münch G, Fulcher I, Lee G, Chau K, Xu B, Kumar R, Hennessy A, Makris A. A pharmacokinetic assessment of optimal dosing, preparation, and chronotherapy of aspirin in pregnancy. Am J Obstet Gynecol 2019; 221:255.e1-255.e9. [PMID: 31051121 DOI: 10.1016/j.ajog.2019.04.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/16/2019] [Accepted: 04/24/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND The benefit of aspirin in preventing preeclampsia is well established; however, studies over the years have demonstrated variability in outcomes with its use. Potential contributing factors to this variation in efficacy include dosing, time of dosing, and preparation of aspirin. OBJECTIVE We aimed to compare the difference in pharmacokinetics of aspirin, through its major active metabolite, salicylic acid, in pregnant women and nonpregnant women, and to examine the effect of dose (100 mg vs 150 mg), preparation (enteric coated vs non-enteric-coated), and chronotherapy of aspirin (morning vs evening) between the 2 groups. MATERIALS AND METHODS Twelve high-risk pregnant women and 3 nonpregnant women were enrolled in this study. Pregnant women were in 1 of 4 groups (100 mg enteric coated, 100 mg non-enteric-coated, 150 mg non-enteric-coated morning dosing, and 150 mg non-enteric-coated evening dosing), whereas nonpregnant women undertook each of the 4 dosing schedules with at least a 30-day washout period. Blood samples were collected at baseline (before ingestion) and at 1, 2, 4, 6, 12, and 24 hours after ingestion of aspirin. Plasma obtained was analyzed for salicylic acid levels by means of liquid chromatography-mass spectrometry. Pharmacokinetic values of area under the curve from time point 0 to 24 hours point of maximum concentration, time of maximum concentration, volume of distribution, clearance, and elimination half-life were analyzed for statistical significance with SPSS v25 software. RESULTS Pregnant women had a 40% ± 4% reduction in area under the curve from time point 0 to 24 hours (P < .01) and 29% ± 3% reduction in point of maximum concentration (P < .01) with a 44% ± 8% increase in clearance (P < .01) in comparison to that in nonpregnant women when 100 mg aspirin was administered. The reduction in the area under the curve from time point 0 to 24 hours, however, was minimized with the use of 150 mg aspirin in pregnant women, with which the area under the curve from time point 0 to 24 hours was closer to that achieved with the use of 100 mg aspirin in nonpregnant women. There was a 4-hour delay (P < .01) in the time of maximum concentration, a 47% ± 3% reduction in point of maximum concentration (P < .01) and a 48% ± 1% increase in volume of distribution (P < .01) with the use of 100 mg enteric-coated aspirin compared to non-enteric-coated aspirin, with no difference in the overall area under the curve. There was no difference in the pharmacokinetics of aspirin between morning and evening dosing. CONCLUSION There is a reduction in the total drug metabolite concentration of aspirin in pregnancy, and therefore a dose adjustment is potentially required in pregnant women. This is likely due to the altered pharmacokinetics of aspirin in pregnancy, with an increase in clearance. There was no difference in the total drug metabolite concentration of aspirin between enteric-coated and non-enteric-coated aspirin and between morning and evening dosing of aspirin. Further pharmacodynamic and clinical studies are required to examine the clinical relevance of these pharmacokinetic findings.
Collapse
Affiliation(s)
- Renuka Shanmugalingam
- School of Medicine, Western Sydney University, NSW, Australia; Department of Renal Medicine, South Western Sydney Local Health District, NSW, Australia; Heart Research Institute, University of Sydney, NSW, Australia; Women's Health Initiative Translational Unit (WHITU), South Western Sydney Local Health District, NSW, Australia.
| | - XiaoSuo Wang
- Bosch Mass Spectrometry Facility, Bosch Institute, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Gerald Münch
- Pharmacology Unit, School of Medicine, Western Sydney University, NSW, Australia
| | - Ian Fulcher
- Department of Obstetrics and Gynaecology, Liverpool Hospital, NSW, Australia
| | - Gaksoo Lee
- Department of Renal Medicine, South Western Sydney Local Health District, NSW, Australia; Women's Health Initiative Translational Unit (WHITU), South Western Sydney Local Health District, NSW, Australia
| | - Katrina Chau
- Heart Research Institute, University of Sydney, NSW, Australia
| | - Bei Xu
- Heart Research Institute, University of Sydney, NSW, Australia
| | - Roshika Kumar
- Department of Obstetrics and Gynaecology, Liverpool Hospital, NSW, Australia
| | - Annemarie Hennessy
- School of Medicine, Western Sydney University, NSW, Australia; Department of Renal Medicine, South Western Sydney Local Health District, NSW, Australia; Heart Research Institute, University of Sydney, NSW, Australia; Women's Health Initiative Translational Unit (WHITU), South Western Sydney Local Health District, NSW, Australia
| | - Angela Makris
- School of Medicine, Western Sydney University, NSW, Australia; Department of Renal Medicine, South Western Sydney Local Health District, NSW, Australia; Heart Research Institute, University of Sydney, NSW, Australia; Women's Health Initiative Translational Unit (WHITU), South Western Sydney Local Health District, NSW, Australia
| |
Collapse
|
122
|
Dallmann A, Ince I, Coboeken K, Eissing T, Hempel G. A Physiologically Based Pharmacokinetic Model for Pregnant Women to Predict the Pharmacokinetics of Drugs Metabolized Via Several Enzymatic Pathways. Clin Pharmacokinet 2019; 57:749-768. [PMID: 28924743 DOI: 10.1007/s40262-017-0594-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Physiologically based pharmacokinetic modeling is considered a valuable tool for predicting pharmacokinetic changes in pregnancy to subsequently guide in-vivo pharmacokinetic trials in pregnant women. The objective of this study was to extend and verify a previously developed physiologically based pharmacokinetic model for pregnant women for the prediction of pharmacokinetics of drugs metabolized via several cytochrome P450 enzymes. METHODS Quantitative information on gestation-specific changes in enzyme activity available in the literature was incorporated in a pregnancy physiologically based pharmacokinetic model and the pharmacokinetics of eight drugs metabolized via one or multiple cytochrome P450 enzymes was predicted. The tested drugs were caffeine, midazolam, nifedipine, metoprolol, ondansetron, granisetron, diazepam, and metronidazole. Pharmacokinetic predictions were evaluated by comparison with in-vivo pharmacokinetic data obtained from the literature. RESULTS The pregnancy physiologically based pharmacokinetic model successfully predicted the pharmacokinetics of all tested drugs. The observed pregnancy-induced pharmacokinetic changes were qualitatively and quantitatively reasonably well predicted for all drugs. Ninety-seven percent of the mean plasma concentrations predicted in pregnant women fell within a twofold error range and 63% within a 1.25-fold error range. For all drugs, the predicted area under the concentration-time curve was within a 1.25-fold error range. CONCLUSION The presented pregnancy physiologically based pharmacokinetic model can quantitatively predict the pharmacokinetics of drugs that are metabolized via one or multiple cytochrome P450 enzymes by integrating prior knowledge of the pregnancy-related effect on these enzymes. This pregnancy physiologically based pharmacokinetic model may thus be used to identify potential exposure changes in pregnant women a priori and to eventually support informed decision making when clinical trials are designed in this special population.
Collapse
Affiliation(s)
- André Dallmann
- Department of Pharmaceutical and Medical Chemistry, Clinical Pharmacy, Westfälische Wilhelms-University Münster, 48149, Münster, Germany.
| | - Ibrahim Ince
- Clinical Pharmacometrics, Bayer AG, 51368, Leverkusen, Germany
| | - Katrin Coboeken
- Clinical Pharmacometrics, Bayer AG, 51368, Leverkusen, Germany
| | - Thomas Eissing
- Clinical Pharmacometrics, Bayer AG, 51368, Leverkusen, Germany
| | - Georg Hempel
- Department of Pharmaceutical and Medical Chemistry, Clinical Pharmacy, Westfälische Wilhelms-University Münster, 48149, Münster, Germany
| |
Collapse
|
123
|
Ramaswamy R, Joshi N, Khan MA, Siddhara S. Nanosomal docetaxel lipid suspension based chemotherapy in a pregnant MBC patient - a case report. Onco Targets Ther 2019; 12:5679-5685. [PMID: 31406465 PMCID: PMC6642620 DOI: 10.2147/ott.s206573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022] Open
Abstract
The current report presents a case of a pregnant woman with breast cancer metastasized to liver and lungs. The standard of care for breast cancer in pregnancy is anthracycline/taxane-based chemotherapy regimens. Docetaxel has shown a favorable toxicity profile during the second and third trimesters of pregnancy. A novel nanosomal docetaxel lipid suspension (NDLS) (DoceAqualip), with a proven efficacy and tolerability profile, has been approved in India for the treatment of advanced solid tumors since 2013. We present here a case of a pregnant woman with metastatic breast cancer managed with NDLS based TAC regimen showing a partial response after six cycles. The patient delivered a healthy male child with normal Apgar score and weight at the 32nd week of gestation.
Collapse
Affiliation(s)
- Rajkumar Ramaswamy
- Department of Medical Oncology, Velammal Medical College and Research Institute, Madurai, Tamil Nadu 625009, India
| | - Nisarg Joshi
- Department of Medical Affairs & Clinical Development, Intas Pharmaceuticals Ltd., Ahmedabad, Gujarat 380054, India
| | - Mujtaba A Khan
- Department of Medical Affairs & Clinical Development, Intas Pharmaceuticals Ltd., Ahmedabad, Gujarat 380054, India
| | - Seerin Siddhara
- Department of Medical Oncology, Velammal Medical College and Research Institute, Madurai, Tamil Nadu 625009, India
| |
Collapse
|
124
|
Patilea-Vrana GI, Unadkat JD. Quantifying Hepatic Enzyme Kinetics of (-)-∆ 9-Tetrahydrocannabinol (THC) and Its Psychoactive Metabolite, 11-OH-THC, through In Vitro Modeling. Drug Metab Dispos 2019; 47:743-752. [PMID: 31048453 PMCID: PMC6556521 DOI: 10.1124/dmd.119.086470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 04/22/2019] [Indexed: 01/21/2023] Open
Abstract
The prevalence of cannabis use and the concentrations of the psychoactive cannabinoid in cannabis, (-)-∆9-tetrahydrocannabinol (THC), are rising. Physiologically based pharmacokinetic modeling and simulations (PBPK M&S) can mechanistically predict exposure of THC and its major and active metabolite, 11-hydroxy-THC (11-OH-THC). To build a THC/11-OH-THC PBPK model, mechanistic information about the disposition of these compounds is necessary, including the drug-metabolizing enzymes (DMEs) involved and the fraction metabolized (fm) and metabolic kinetic parameters (intrinsic clearance, maximal formation rate, and Km) via the identified enzymes. We previously identified and quantified the fm of DMEs involved in hepatic depletion of THC and 11-OH-THC. In this study, we extend this work to characterize the enzyme kinetics of THC and 11-OH-THC by monitoring their depletion and formation of some of their metabolites in pooled human liver microsomes. A P450 and UDP-glucuronosyltransferase (UGT) kinetic model was fitted to the concentration-time depletion/formation profiles to establish the contribution and kinetics of the individual DME pathways. CYP2C9 pathway was the major pathway for depletion of THC (fm = 0.91, Km,u = 3 nM) and formation of 11-OH-THC. The remaining THC depletion pathway was attributed to CYP2D6. 11-OH-THC was depleted by UGTs (fm = 0.67 and Km,u = 39 nM), CYP3A4 (fm = 0.18, Km,u = 824 nM), and CYP2C9 (fm = 0.15, Km,u = 33 nM). These mechanistic in vitro data can be used to predict the exposure of THC and 11-OH-THC in healthy and special populations, including in the presence of drug-drug interactions, via PBPK M&S.
Collapse
Affiliation(s)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
125
|
Advancing the development of new tuberculosis treatment regimens: The essential role of translational and clinical pharmacology and microbiology. PLoS Med 2019; 16:e1002842. [PMID: 31276490 PMCID: PMC6611566 DOI: 10.1371/journal.pmed.1002842] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
126
|
Abstract
Medicine use in pregnancy is extremely common, but there are significant knowledge gaps surrounding the safety, dosage and long-term effects of drugs used. Pregnant women have been purposively excluded from clinical trials of the majority of treatments for conditions that may occur concurrently with pregnancy. There is minimal information on the pharmacokinetics of many existing treatments and no systematic capture of long-term outcome data to help inform choices. Treatments commonly used in pregnancy are thus often old and untested, not optimised in dose, and prescribed off-label without adequate safety information. In addition, there has been a staggering lack of investment in drug development for obstetric conditions for decades. This is a major public health concern, and pregnancy complications are the leading cause of mortality in children under five years old globally, and health in pregnancy is a major determinant of women's long-term health and wellbeing. There is an acute need for adequate investment and legislation to boost inclusion of pregnant women in clinical studies, capture high-quality information on medication use in pregnancy in general, and encourage new medicinal product development for obstetric conditions.
Collapse
Affiliation(s)
- Sarah JE Stock
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Nine Edinburgh BioQuarter, 9 Little France Road, Edinburgh, EH16 4UX, UK
| | - Jane E Norman
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh BioQuarter, Edinburgh, EH16 4SA, UK
- Faculty of Health Sciences, University of Bristol, 5 Tyndall Avenue, Bristol, UK
| |
Collapse
|
127
|
Shah M, Xu M, Shah P, Wang X, Clark SM, Costantine M, West HA, Nanovskaya TN, Ahmed MS, Abdel-Rahman SZ, Venkataramanan R, Caritis SN, Hankins GDV, Rytting E. Effect of CYP2C9 Polymorphisms on the Pharmacokinetics of Indomethacin During Pregnancy. Eur J Drug Metab Pharmacokinet 2019; 44:83-89. [PMID: 30159654 DOI: 10.1007/s13318-018-0505-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND OBJECTIVE Cytochrome P450 (CYP) 2C9 catalyzes the biotransformation of indomethacin to its inactive metabolite O-desmethylindomethacin (DMI). The aim of this work was to determine the effect of CYP2C9 polymorphisms on indomethacin metabolism in pregnant women. METHODS Plasma concentrations of indomethacin and DMI at steady state were analyzed with a validated LC-MS/MS method. DNA was isolated from subject blood and buccal smear samples. Subjects were grouped by genotype for comparisons of pharmacokinetic parameters. RESULTS For subjects with the *1/*2 genotype, the mean steady-state apparent oral clearance (CL/Fss) of indomethacin was 13.5 ± 7.7 L/h (n = 4) and the mean metabolic ratio (AUCDMI/AUCindomethacin) was 0.291 ± 0.133. For subjects with the *1/*1 genotype, these values were 12.4 ± 2.7 L/h and 0.221 ± 0.078, respectively (n = 14). Of note, we identified one subject who was a carrier of both the *3 and *4 alleles, resulting in an amino acid change (I359P) which has not been reported previously. This subject had a metabolic ratio of 0.390 and a CL/Fss of indomethacin (24.3 L/h) that was nearly double the wild-type clearance. CONCLUSION Although our results are limited by sample size and are not statistically significant, these data suggest that certain genetic polymorphisms of CYP2C9 may lead to an increased metabolic ratio and an increase in the clearance of indomethacin. More data are needed to assess the impact of CYP2C9 genotype on the effectiveness of indomethacin as a tocolytic agent.
Collapse
Affiliation(s)
- Mansi Shah
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Meixiang Xu
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Poonam Shah
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Xiaoming Wang
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Shannon M Clark
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Maged Costantine
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Holly A West
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Tatiana N Nanovskaya
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mahmoud S Ahmed
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sherif Z Abdel-Rahman
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Raman Venkataramanan
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Steve N Caritis
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Gary D V Hankins
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Erik Rytting
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
128
|
Abstract
: Product labels for cobicistat with atazanavir or darunavir, and for elvitegravir/cobicistat/emtricitabine/tenofovir (alafenamide or disoproxil fumarate) were recently updated to state that these products are not recommended for initiation during pregnancy, and an alternative regimen is recommended for those who become pregnant during therapy with these products. Herein, we present the rationale for these recommendations, which are based on studies in pregnant women evaluating the pharmacokinetics and antiviral activity of darunavir/cobicistat or elvitegravir/cobicistat-containing antiretroviral regimens. In these studies, mean steady-state minimum concentrations in the second and third trimester versus postpartum of cobicistat, darunavir, and elvitegravir were reduced by 61-83%, 89-92%, and 82-86%, respectively. In the absence of data with atazanavir/cobicistat, we leveraged the available data with darunavir/cobicistat and elvitegravir/cobicistat to make recommendations for atazanavir/cobicistat. Darunavir/ritonavir and atazanavir/ritonavir remain viable treatment options for pregnant women.
Collapse
|
129
|
Abstract
BACKGROUND Antiepileptic drugs (AEDs) are the mainstay of epilepsy treatment. Since 1989, 18 new AEDs have been licensed for clinical use and there are now 27 licensed AEDs in total for the treatment of patients with epilepsy. Furthermore, several AEDs are also used for the management of other medical conditions, for example, pain and bipolar disorder. This has led to an increasingly widespread application of therapeutic drug monitoring (TDM) of AEDs, making AEDs among the most common medications for which TDM is performed. The aim of this review is to provide an overview of the indications for AED TDM, to provide key information for each individual AED in terms of the drug's prescribing indications, key pharmacokinetic characteristics, associated drug-drug pharmacokinetic interactions, and the value and the intricacies of TDM for each AED. The concept of the reference range is discussed as well as practical issues such as choice of sample types (total versus free concentrations in blood versus saliva) and sample collection and processing. METHODS The present review is based on published articles and searches in PubMed and Google Scholar, last searched in March 2018, in addition to references from relevant articles. RESULTS In total, 171 relevant references were identified and used to prepare this review. CONCLUSIONS TDM provides a pragmatic approach to epilepsy care, in that bespoke dose adjustments are undertaken based on drug concentrations so as to optimize clinical outcome. For the older first-generation AEDs (carbamazepine, ethosuximide, phenobarbital, phenytoin, primidone, and valproic acid), much data have accumulated in this regard. However, this is occurring increasingly for the new AEDs (brivaracetam, eslicarbazepine acetate, felbamate, gabapentin, lacosamide, lamotrigine, levetiracetam, oxcarbazepine, perampanel, piracetam, pregabalin, rufinamide, stiripentol, sulthiame, tiagabine, topiramate, vigabatrin, and zonisamide).
Collapse
|
130
|
Huang Z, Pan X, Zhou J, Leung WT, Li C, Wang L. Chinese herbal medicine for acute upper respiratory tract infections and reproductive safety: A systematic review. Biosci Trends 2019; 13:117-129. [PMID: 30930358 DOI: 10.5582/bst.2018.01298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Acute upper respiratory tract infections (AURTIs) are common and self-limited in people with normal immunity but sometimes lead to poor clinical outcomes under specific conditions such as pregnancy if not treated appropriately. Chinese herbal medicines (CHM), which are widely used to treat AURTIs, have proven to be effective in preclinical and clinical studies. This review focuses on the bioactivities of typical CHM and the adverse reactions they cause, and especially issues with reproductive safety when treating AURTIs. The main mechanisms for clinical efficacy may include anti-viral, anti-bacterial, anti-inflammatory, antipyretic, and immunomodulatory action as indicated by preclinical evidence. Most clinical trials indicate that CHM shortens the natural course of AURTIs and that it relieves related symptoms such as a fever, headaches, coughing, myalgia, a cold, sore throat, and a nasal obstruction. However, some CHM have a range of adverse effects and potentially affect reproduction from endocrinal secretion to embryo development while others do not. Therefore, clinical adverse reactions and preclinical studies on the toxicity of CHM are discussed. More reliable evidence is required to conclude that CHM are efficacious and safe for pregnant women with AURTIs. This review should help to promote advances in the research on and development of CHM as alternative treatments for AURTIs and offer insight into strategies to manage the safety of CHM during clinical use.
Collapse
Affiliation(s)
- Zengshu Huang
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Xinyao Pan
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Jing Zhou
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Wing Ting Leung
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Chuyu Li
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| | - Ling Wang
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Shanghai Medical College, Fudan University.,The Academy of Integrative Medicine, Fudan University.,Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
| |
Collapse
|
131
|
Crauwels HM, Osiyemi O, Zorrilla C, Bicer C, Brown K. Reduced exposure to darunavir and cobicistat in HIV-1-infected pregnant women receiving a darunavir/cobicistat-based regimen. HIV Med 2019; 20:337-343. [PMID: 30873741 DOI: 10.1111/hiv.12721] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate darunavir and cobicistat pharmacokinetics in pregnant women with HIV-1 infection. METHODS This phase 3b, open-label study enrolled HIV-1-infected pregnant women (18-26 weeks of gestation) receiving combination antiretroviral therapy with once-daily darunavir/cobicistat 800/150 mg. The plasma pharmacokinetics of darunavir (total and unbound) and cobicistat were assessed over 24 h during the second and third trimesters (24-28 and 34-38 weeks of gestation, respectively) and 6-12 weeks postpartum. Pharmacokinetic parameters [area under the plasma concentration-time curve over 24 h (AUC24 h ), maximum plasma concentration (Cmax ) and minimum plasma concentration (Cmin )] were derived using noncompartmental analysis and compared using linear mixed effects modelling (pregnancy versus postpartum). Antiviral activity and safety were evaluated. RESULTS Seven women were enrolled in the study; six completed it. Total darunavir exposure was lower during pregnancy than postpartum (AUC24 h , 50-56% lower; Cmax , 37-49% lower; Cmin , 89-92% lower); unbound darunavir exposure was also reduced (AUC24 h , 40-45% lower; Cmax , 32-41% lower; Cmin , 88-92% lower). Cobicistat exposure was also lower during pregnancy than postpartum (AUC24 h , 49-63% lower; Cmax , 27-50% lower; Cmin , 83% lower). At study completion, five of six (83%) women were virologically suppressed (HIV-1 RNA < 50 copies/mL). There was one virological failure (the patient was nonadherent; no emerging genotypic resistance was observed and susceptibility to antiretrovirals was maintained). No mother-to-child transmission was detected among six infants born to the six women who completed the study. Overall, darunavir/cobicistat was well tolerated in women and infants. CONCLUSIONS In view of markedly reduced darunavir and cobicistat exposures during pregnancy, this combination is not recommended in HIV-1-infected pregnant women.
Collapse
Affiliation(s)
| | - O Osiyemi
- Triple O Research Institute PA, West Palm Beach, FL, USA
| | - C Zorrilla
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - C Bicer
- BICER Consulting & Research, Antwerp, Belgium
| | - K Brown
- Janssen Research & Development, LLC, Titusville, NJ, USA
| |
Collapse
|
132
|
Biesdorf C, Martins FS, Sy SKB, Diniz A. Physiologically-based pharmacokinetics of ziprasidone in pregnant women. Br J Clin Pharmacol 2019; 85:914-923. [PMID: 30669177 DOI: 10.1111/bcp.13872] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/29/2018] [Accepted: 01/06/2019] [Indexed: 01/19/2023] Open
Abstract
AIMS Pregnancy is associated with physiological changes that alter the pharmacokinetics (PK) of drugs. The aim of this study was to predict the PK of ziprasidone in pregnant women. METHODS A full physiologically-based pharmacokinetic (PBPK) model of ziprasidone was developed and validated for the non-pregnant population (healthy adults, paediatrics, geriatrics), and this was extended to the pregnant state to assess the change in PK profile of ziprasidone throughout pregnancy. RESULTS The PBPK model successfully predicted the ziprasidone disposition in healthy adult volunteers, wherein the predicted and observed AUC, Cmax and tmax were within the fold-difference of 0.94-1.09, 0.89-1.40 and 0.80-1.08, respectively. The paediatric and geriatric population, also showed predicted AUC, Cmax and tmax within a two-fold range of the observed values. The simulated exposure in pregnant women using a p-PBPK model showed no significant difference when compared to non-pregnant women. CONCLUSIONS The PBPK model predicted the impact of physiological changes during pregnancy on PK and exposure of ziprasidone, suggesting that dose adjustment is not necessary in this special population.
Collapse
Affiliation(s)
- Carla Biesdorf
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| | | | - Sherwin K B Sy
- Department of Statistics, State University of Maringá, Maringá, Brazil
| | - Andrea Diniz
- Department of Pharmacy, State University of Maringá, Maringá, Brazil
| |
Collapse
|
133
|
Birgersson S, Valea I, Tinto H, Traore-Coulibaly M, Toe LC, Hoglund RM, Van Geertruyden JP, Ward SA, D’Alessandro U, Abelö A, Tarning J. Population pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant women with uncomplicated Plasmodium falciparum malaria in Burkina Faso: an open label trial. Wellcome Open Res 2019; 4:45. [PMID: 32025570 PMCID: PMC6974929 DOI: 10.12688/wellcomeopenres.14849.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Malaria during pregnancy is a major health risk for both the mother and the foetus. Pregnancy has been shown to influence the pharmacokinetics of a number of different antimalarial drugs. This might lead to an under-exposure in these patients which could increase the risk of treatment failure and the development of drug resistance. The study aim was to evaluate the pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant patients using a population modelling approach. Methods: Twenty-four women in their second and third trimester of pregnancy and twenty-four paired non-pregnant women, all with uncomplicated P. falciparum malaria, were enrolled in this study. Treatment was a fixed-dose combination of oral artesunate and mefloquine once daily for three days. Frequent blood samples were collected and concentration-time data for artesunate and dihydroartemisinin were analysed simultaneously using nonlinear mixed-effects modelling. Results: Artesunate pharmacokinetics was best described by a transit-compartment absorption model followed by a one-compartment disposition model under the assumption of complete in vivo conversion of artesunate into dihydroartemisinin. Dihydroartemisinin pharmacokinetics was best described by a one-compartment disposition model with first-order elimination. Pregnant women had a 21% higher elimination clearance of dihydroartemisinin, compared to non-pregnant women, resulting in proportionally lower drug exposure. In addition, initial parasitaemia and liver status (alanine aminotransferase) were found to affect the relative bioavailability of artesunate. Conclusions: Results presented here show a substantially lower drug exposure to the antimalarial drug dihydroartemisinin during pregnancy after standard oral treatment of artesunate and mefloquine. This might result in an increased risk of treatment failure and drug resistance development, especially in low transmission settings where relative immunity is lower. Trial registration: ClinicalTrials.gov NCT00701961 (19/06/2008).
Collapse
Affiliation(s)
- Sofia Birgersson
- Department of Pharmacology, University of Gothenburg, Gothenburg, 405 30, Sweden
| | - Innocent Valea
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
- Institut de Recherche en Sciences de la Sante´, Unite´ de Recherche Clinique de Nanoro, Nanoro, Burkina Faso
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
- Institut de Recherche en Sciences de la Sante´, Unite´ de Recherche Clinique de Nanoro, Nanoro, Burkina Faso
| | - Maminata Traore-Coulibaly
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
- Institut de Recherche en Sciences de la Sante´, Unite´ de Recherche Clinique de Nanoro, Nanoro, Burkina Faso
| | - Laeticia C. Toe
- Institut de Recherche en Sciences de la Santé, Direction Régionale de l’Ouest Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
- Department of Food Safety, Quality and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, B-9000, Belgium
| | - Richard M. Hoglund
- Mahidol-Oxford Tropical Medicine Resarch Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | | | - Stephen A. Ward
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | | | - Angela Abelö
- Department of Pharmacology, University of Gothenburg, Gothenburg, 405 30, Sweden
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Resarch Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| |
Collapse
|
134
|
Birgersson S, Valea I, Tinto H, Traore-Coulibaly M, Toe LC, Hoglund RM, Van Geertruyden JP, Ward SA, D’Alessandro U, Abelö A, Tarning J. Population pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant women with uncomplicated Plasmodium falciparum malaria in Burkina Faso: an open label trial. Wellcome Open Res 2019. [DOI: 10.12688/wellcomeopenres.14849.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Malaria during pregnancy is a major health risk for both the mother and the foetus. Pregnancy has been shown to influence the pharmacokinetics of a number of different antimalarial drugs. This might lead to an under-exposure in these patients which could increase the risk of treatment failure and the development of drug resistance. The study aim was to evaluate the pharmacokinetics of artesunate and dihydroartemisinin in pregnant and non-pregnant patients using a population modelling approach. Methods: Twenty-four women in their second and third trimester of pregnancy and twenty-four paired non-pregnant women, all with uncomplicated P. falciparum malaria, were enrolled in this study. Treatment was a fixed-dose combination of oral artesunate and mefloquine once daily for three days. Frequent blood samples were collected and concentration-time data for artesunate and dihydroartemisinin were analysed simultaneously using nonlinear mixed-effects modelling. Results: Artesunate pharmacokinetics was best described by a transit-compartment absorption model followed by a one-compartment disposition model under the assumption of complete in vivo conversion of artesunate into dihydroartemisinin. Dihydroartemisinin pharmacokinetics was best described by a one-compartment disposition model with first-order elimination. Pregnant women had a 21% higher elimination clearance of dihydroartemisinin, compared to non-pregnant women, resulting in proportionally lower drug exposure. In addition, initial parasitaemia and liver status (alanine aminotransferase) were found to affect the relative bioavailability of artesunate. Conclusions: Results presented here show a substantially lower drug exposure to the antimalarial drug dihydroartemisinin during pregnancy after standard oral treatment of artesunate and mefloquine. This might result in an increased risk of treatment failure and drug resistance development, especially in low transmission settings where relative immunity is lower. Trial registration: ClinicalTrials.gov NCT00701961 (19/06/2008)
Collapse
|
135
|
Patilea-Vrana GI, Anoshchenko O, Unadkat JD. Hepatic Enzymes Relevant to the Disposition of (-)-∆ 9-Tetrahydrocannabinol (THC) and Its Psychoactive Metabolite, 11-OH-THC. Drug Metab Dispos 2019; 47:249-256. [PMID: 30567877 PMCID: PMC6374540 DOI: 10.1124/dmd.118.085548] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/14/2018] [Indexed: 01/03/2023] Open
Abstract
Marijuana use by pregnant women is increasing. To predict developmental risk to the fetus/neonate from such use, in utero fetal exposure to (-)-∆9-tetrahydrocannabinol (THC), the main psychoactive cannabinoid in marijuana and its active psychoactive metabolite, 11-hydroxy-∆9-tetrahydrocannabinol (11-OH-THC), needs to be determined. Since such measurement is not possible, physiologically based pharmacokinetic (PBPK) modeling and simulation can provide an alternative method to estimate fetal exposure to cannabinoids. To do so, pharmacokinetic parameters for the disposition of THC and 11-OH-THC need to be elucidated. Here, we report a first step to estimate these parameters, namely, those related to maternal metabolism of THC/11-OH-THC in human liver microsomes (HLMs) at plasma concentrations observed after smoking marijuana. Using recombinant cytochrome P450 (P450) and UDP-glucuronosyltransferase (UGT) enzymes, CYP1A1, 1A2, 2C9, 2C19, 2D6, 3A4, 3A5, 3A7, and UGT1A9 and UGT2B7 were found to be involved in the disposition of THC/11-OH-THC. Using pooled HLMs, the fraction metabolized (f m) by relevant enzymes was measured using selective enzyme inhibitors, and then adjusted for enzyme cross-inhibition. As previously reported, CYP2C9 was the major enzyme responsible for depletion of THC and formation of 11-OH-THC with f m values of 0.82 ± 0.08 and 0.99 ± 0.10, respectively (mean ± S.D.), while CYP2D6 and CYP2C19 were minor contributors. 11-OH-THC was depleted by UGT and P450 enzymes with f m values of 0.60 ± 0.05 and 0.40 ± 0.05, respectively (mean ± S.D.), with UGT2B7, UGT1A9, CYP2C9, and CYP3A4 as contributors. These mechanistic data represent the first set of drug-dependent parameters necessary to predict maternal-fetal cannabinoid exposure during pregnancy using PBPK modeling.
Collapse
Affiliation(s)
| | - Olena Anoshchenko
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington
| |
Collapse
|
136
|
Gini J, Olagunju A, Dickinson L, Waitt C, Neary M, Else LJ, Siccardi M, Khoo S. Impact of pharmacogenetics and pregnancy on tenofovir and emtricitabine pharmacokinetics. Pharmacogenomics 2019; 20:217-223. [PMID: 30767719 DOI: 10.2217/pgs-2018-0111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM Treatment and prevention of mother-to-child transmission of HIV in pregnancy utilizes tenofovir (TFV) and emtricitabine (FTC) as NRTI backbone in combination with a third agent from a different class. We hypothesized that combined effect of pregnancy and pharmacogenetics significantly changes TFV and FTC pharmacokinetics (PK). Therefore, this study aims to evaluate the role of SNPs of transporters (ABCC2 and ABCC4) on TFV and FTC PK during pregnancy. METHOD 61 pregnant or postpartum women on TFV and FTC were selected from a group of pregnant and postpartum Nigerian women and both SNPs and drug levels were evaluated. RESULTS Pregnancy decreases TFV plasma concentration by 26% (log10 β = -0.131 [-0.228, -0.034; p = 0.009] at median [range] time-point postdose 14 [7-18.5h]). FTC concentration in individuals with ABCC2 12:g.154962860T>C TT genotype were one- to twofold higher than heterozygous (CT) and homozygous (CC) women. All other evaluated SNPs were not significant. CONCLUSION Pregnancy decreased TFV concentration and significant relationship was found between FTC and ABCC2 12:g.154962860T>C wild-type allele. However, the interplay between pregnancy and pharmacogenetics on TFV and FTC PK is unclear but require further evaluation.
Collapse
Affiliation(s)
- Joshua Gini
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Adeniyi Olagunju
- Faculty of Pharmacy, Obafemi Awolowo University Ile-Ife, Osun State, Nigeria
| | - Laura Dickinson
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Catriona Waitt
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK.,Royal Liverpool University Hospital, Tropical and Infectious diseases department, Liverpool, UK
| | - Megan Neary
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Laura J Else
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Marco Siccardi
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Saye Khoo
- Department of Molecular & Clinical Pharmacology, University of Liverpool, Liverpool, UK.,Royal Liverpool University Hospital, Tropical and Infectious diseases department, Liverpool, UK
| |
Collapse
|
137
|
Analysis of bupivacaine enantiomers in plasma as total and unbound concentrations using LC-MS/MS: Application in a pharmacokinetic study of a parturient with placental transfer. J Pharm Biomed Anal 2019; 164:268-275. [DOI: 10.1016/j.jpba.2018.10.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/17/2018] [Accepted: 10/21/2018] [Indexed: 01/07/2023]
|
138
|
Abstract
Purpose of review Antipsychotics are frequently prescribed to women of childbearing age and are increasingly prescribed during pregnancy. A small, but growing, body of research on implications for pregnancy and infant outcomes is available to inform the risks and benefits of in utero exposure to antipsychotics. This review examines the existing published research on the use of common typical and atypical antipsychotics in pregnancy and the implications for pregnancy and infant outcomes. Recent findings The majority of studies do not show associations with major malformations and antipsychotic use in pregnancy, with the possible exception of risperidone. There is concern that atypical antipsychotics may be associated with gestational diabetes. Metabolic changes during pregnancy may necessitate dose adjustments. Summary In general, it is recommended that women who need to take an antipsychotic during pregnancy continue the antipsychotic that has been most effective for symptom remission. Further study on risperidone is needed to better understand its association with malformations and it is not considered a first-line agent for use during pregnancy.
Collapse
Affiliation(s)
- Hannah K Betcher
- Department of Psychiatry, Northwestern University Feinberg School of Medicine 676 N. St. Clair St. Ste 1000, Chicago, IL 60611, USA.,Mayo Clinic, Rochester, MN, USA
| | - Catalina Montiel
- Department of Psychiatry, Northwestern University Feinberg School of Medicine 676 N. St. Clair St. Ste 1000, Chicago, IL 60611, USA
| | - Crystal T Clark
- Department of Psychiatry, Northwestern University Feinberg School of Medicine 676 N. St. Clair St. Ste 1000, Chicago, IL 60611, USA.,Department of Obstetrics and Gynecology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
139
|
van Donge T, Evers K, Koch G, van den Anker J, Pfister M. Clinical Pharmacology and Pharmacometrics to Better Understand Physiological Changes During Pregnancy and Neonatal Life. Handb Exp Pharmacol 2019; 261:325-337. [PMID: 30968215 DOI: 10.1007/164_2019_210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pregnant women, fetuses, and newborns are particularly vulnerable patient populations. During pregnancy, the body is subject to physiological changes that influence the pharmacokinetics and pharmacodynamics of drugs. Inappropriate dosing in pregnant women can result in sub-therapeutic or toxic effects, putting not only the pregnant woman but also her fetus at risk. During neonatal life, maturation processes also affect pharmacokinetics and pharmacodynamics of drugs. Inappropriate dosing in newborns leads not only to short-term complications but can also have a negative impact on the long-term development of infants and children. For these reasons, it is crucial to characterize physiological changes in pregnant women, describe placental transfer kinetics of drugs, and describe physiological changes related to the transition from intrauterine to extrauterine life and maturation processes in preterm and term neonates. Quantitative pharmacological approaches such as pharmacometric and physiologically-based modeling and model-based simulations can be useful to better understand and predict such physiological changes and their effects on drug exposure and response. This review article (1) gives an overview of physiological changes in pregnant women, their fetuses, and (pre)term neonates, (2) presents case studies to illustrate applications of new modeling and simulation approaches, and (3) discusses challenges and opportunities in optimizing and personalizing treatments during pregnancy and neonatal life.
Collapse
Affiliation(s)
- Tamara van Donge
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.
| | - Katrina Evers
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Gilbert Koch
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| | - Marc Pfister
- Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland.,Certara LP, Princeton, NJ, USA
| |
Collapse
|
140
|
Barzilai M, Avivi I, Amit O. Hematological malignancies during pregnancy. Mol Clin Oncol 2018; 10:3-9. [PMID: 30655971 DOI: 10.3892/mco.2018.1759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 06/26/2018] [Indexed: 12/27/2022] Open
Abstract
Hematological malignancy during pregnancy is a rare event, therefore most data on this issue is based on case studies, retrospective studies and expert opinion. The purpose of the present narrative review was to provide an overview of the diagnosis and recommended management of the most common hematological malignancies during pregnancy, based on current literature, with clinical cases, and discussion of the diagnostic and therapeutic options. The therapeutic consensus while coping with hematological malignancies in pregnancy is to salvage the mother, while trying to preserve pregnancy and avoid treatment-related-toxicity to the fetus. In most scenarios, particularly during late trimesters, the goal is to administer the same treatment as outside of pregnancy, if possible. Further research is needed for better evidence-based management.
Collapse
Affiliation(s)
- Merav Barzilai
- Hematology and Hemato-Oncology Division, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Irit Avivi
- Hematology and Hemato-Oncology Division, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Odelia Amit
- Hematology and Hemato-Oncology Division, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| |
Collapse
|
141
|
Carvalho DM, Lanchote VL, Filgueira GCDO, Nardotto GHB, Duarte G, Cavalli RC, Moisés ECD. Pharmacokinetics and Transplacental Transfer of Fluoxetine Enantiomers and Their Metabolites in Pregnant Women. Clin Pharmacol Ther 2018; 105:1003-1008. [PMID: 30346625 DOI: 10.1002/cpt.1263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 09/29/2018] [Indexed: 11/07/2022]
Abstract
Considering that fluoxetine (FLX) is used to treat depressive states during pregnancy and that it is a cytochrome P450 (CYP)2D6 inhibitor, which is involved in the metabolism of both of its enantiomers, this study aims to describe the enantioselective distribution and metabolism of FLX and of its metabolite norfluoxetine (NorFLX) following a single oral dose. Nine healthy pregnant women received 20 mg FLX at 32 weeks of gestation and later at the day of delivery. The apparent clearance of (S)-(+)-FLX (1.45 vs. 0.66 L/hour/kg) and the area under the plasma concentration vs. time curve (AUC) of the (S)-(+)-NorFLX (AUC0-∞ 942.7 vs. 498.6 ng hour/mL) were higher (P < 0.05) than those of the respective (R)-(-) enantiomers, indicating that the (S)-(+)-FLX enantiomer is preferentially metabolized to (S)-(+)-NorFLX. The placental transfer (umbilical vein/maternal vein) of FLX and NorFLX is low (30-40%), with the predominant transfer of (S)-(+)-FLX (44 vs. 33%). The distribution of the enantiomers of FLX and NorFLX to amniotic fluid is low (< 10%).
Collapse
Affiliation(s)
- Daniela Miarelli Carvalho
- 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, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Glauco Henrique Balthazar Nardotto
- Department of Clinical, Toxicologic and Bromatologic Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, 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
| | - Ricardo Carvalho Cavalli
- Department of Obstetrics and Gynecology, Ribeirão Preto Medical School, 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
| |
Collapse
|
142
|
Speed V, Roberts LN, Patel JP, Arya R. Venous thromboembolism and women's health. Br J Haematol 2018; 183:346-363. [DOI: 10.1111/bjh.15608] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Victoria Speed
- King's Thrombosis Centre; Department of Haematological Medicine; King's College Hospital NHS Foundation Trust; London UK
- Institute of Pharmaceutical Sciences; King's College London; London UK
| | - Lara N. Roberts
- King's Thrombosis Centre; Department of Haematological Medicine; King's College Hospital NHS Foundation Trust; London UK
| | - Jignesh P. Patel
- King's Thrombosis Centre; Department of Haematological Medicine; King's College Hospital NHS Foundation Trust; London UK
- Institute of Pharmaceutical Sciences; King's College London; London UK
| | - Roopen Arya
- King's Thrombosis Centre; Department of Haematological Medicine; King's College Hospital NHS Foundation Trust; London UK
| |
Collapse
|
143
|
Population Pharmacokinetics of Artemether, Dihydroartemisinin, and Lumefantrine in Rwandese Pregnant Women Treated for Uncomplicated Plasmodium falciparum Malaria. Antimicrob Agents Chemother 2018; 62:AAC.00518-18. [PMID: 30061282 PMCID: PMC6153812 DOI: 10.1128/aac.00518-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
The artemisinin-based combination therapy artemether-lumefantrine is commonly used in pregnant malaria patients. However, the effect of pregnancy-related changes on exposure is unclear, and pregnancy has been associated with decreased efficacy in previous studies. This study aimed to characterize the population pharmacokinetics of artemether, its active metabolite dihydroartemisinin, and lumefantrine in 22 Rwandese pregnant women in their second (n = 11) or third (n = 11) trimester with uncomplicated Plasmodium falciparum malaria. These patients were enrolled from Rwamagana district hospital and received the standard fixed oral dose combination of 80 mg of artemether and 480 mg of lumefantrine twice daily for 3 days. Venous plasma concentrations were quantified for all three analytes using liquid chromatography coupled with tandem mass spectroscopy, and data were analyzed using nonlinear mixed-effects modeling. Lumefantrine pharmacokinetics was described by a flexible but highly variable absorption, with a mean absorption time of 4.04 h, followed by a biphasic disposition model. The median area under the concentration-time curve from 0 h to infinity (AUC0-∞) for lumefantrine was 641 h · mg/liter. Model-based simulations indicated that 11.7% of the study population did not attain the target day 7 plasma concentration (280 ng/ml), a threshold associated with increased risk of recrudescence. The pharmacokinetics of artemether was time dependent, and the autoinduction of its clearance was described using an enzyme turnover model. The turnover half-life was predicted to be 30.4 h. The typical oral clearance, which started at 467 liters/h, increased 1.43-fold at the end of treatment. Simulations suggested that lumefantrine pharmacokinetic target attainment appeared to be reassuring in Rwandese pregnant women, particularly compared to target attainment in Southeast Asia. Larger cohorts will be required to confirm this finding.
Collapse
|
144
|
Regitz-Zagrosek V, Roos-Hesselink JW, Bauersachs J, Blomström-Lundqvist C, Cífková R, De Bonis M, Iung B, Johnson MR, Kintscher U, Kranke P, Lang IM, Morais J, Pieper PG, Presbitero P, Price S, Rosano GMC, Seeland U, Simoncini T, Swan L, Warnes CA. 2018 ESC Guidelines for the management of cardiovascular diseases during pregnancy. Eur Heart J 2018; 39:3165-3241. [PMID: 30165544 DOI: 10.1093/eurheartj/ehy340] [Citation(s) in RCA: 1269] [Impact Index Per Article: 181.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
145
|
Zelop CM, Einav S, Mhyre JM, Martin S. Cardiac arrest during pregnancy: ongoing clinical conundrum. Am J Obstet Gynecol 2018; 219:52-61. [PMID: 29305251 DOI: 10.1016/j.ajog.2017.12.232] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/16/2017] [Accepted: 12/27/2017] [Indexed: 02/03/2023]
Abstract
While global maternal mortality has decreased in the last 25 years, the maternal mortality ratio in the United States has actually increased. Maternal mortality is a complex phenomenon involving multifaceted socioeconomic and clinical parameters including inequalities in access to health care, racial and ethnic disparities, maternal comorbidities, and epidemiologic ascertainment bias. Escalating maternal mortality underscores the importance of clinician preparedness to respond to maternal cardiac arrest that may occur in any maternal health care setting. Management of maternal cardiac arrest requires an interdisciplinary team familiar with the physiologic changes of pregnancy and the maternal resuscitation algorithm. Interventions intended to mitigate obstacles such as aortocaval compression, which may undermine the success of resuscitation interventions, must be performed concurrent to standard basic and advanced cardiac life support maneuvers. High-quality chest compressions and oxygenation must be performed along with manual left lateral uterine displacement when the uterine size is ≥20 weeks. While deciphering the etiology of maternal cardiac arrest, diagnoses unique to pregnancy and those of the nonpregnant state should be considered at the same time. If initial basic life support and advanced cardiac life support interventions fail to restore maternal circulation within 4 minutes of cardiac arrest, perimortem delivery is advised provided the uterus is ≥20 weeks' size. Preparations for perimortem delivery are best anticipated by the resuscitation team for the procedure to be executed opportunely. Following delivery, intraabdominal examination may reveal a vascular catastrophe, hematoma, or both. If return of spontaneous circulation has not been achieved, additional interventions may include cardiopulmonary bypass and/or extracorporeal membrane oxygenation. Simulation and team training enhance institution readiness for maternal cardiac arrest. Knowledge gaps are significant in the science of maternal resuscitation. Further research is required to fully optimize: relief of aortocaval compression during the resuscitation process, gestational age and timing of perimortem delivery, and other interventions that deviate from nonpregnant standard resuscitation protocol to achieve successful maternal resuscitation. A robust detailed national and international prospective database was recommended by the International Liaison Committee on Resuscitation in 2015 to facilitate further research unique to cardiac arrest during pregnancy that will produce optimal resuscitation techniques for maternal cardiac arrest.
Collapse
|
146
|
Tong M, Stanley JL, Chen Q, James JL, Stone PR, Chamley LW. Placental Nano-vesicles Target to Specific Organs and Modulate Vascular Tone In Vivo. Hum Reprod 2018; 32:2188-2198. [PMID: 29040541 DOI: 10.1093/humrep/dex310] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 10/05/2017] [Indexed: 01/15/2023] Open
Abstract
STUDY QUESTION How do nano-vesicles extruded from normal first trimester human placentae affect maternal vascular function? SUMMARY ANSWER Placental nano-vesicles affect the ability of systemic mesenteric arteries to undergo endothelium- and nitric oxide- (NO-) dependent vasodilation in vivo in pregnant mice. WHAT IS KNOWN ALREADY Dramatic cardiovascular adaptations occur during human pregnancy, including a substantial decrease in total peripheral resistance in the first trimester. The human placenta constantly extrudes extracellular vesicles that can enter the maternal circulation and these vesicles may play an important role in feto-maternal communication. STUDY DESIGN, SIZE, DURATION Human placental nano-vesicles were administered into CD1 mice via a tail vein and their localization and vascular effects at 30 min and 24 h post-injection were investigated. PARTICIPANTS/MATERIALS, SETTING, METHODS Nano-vesicles from normal first trimester human placentae were collected and administered into pregnant (D12.5) or non-pregnant female mice. After either 30 min or 24 h of exposure, all major organs were dissected for imaging (n = 7 at each time point) while uterine and mesenteric arteries were dissected for wire myography (n = 6 at each time point). Additional in vitro studies using HMEC-1 endothelial cells were also conducted to investigate the kinetics of interaction between placental nano-vesicles and endothelial cells. MAIN RESULTS AND THE ROLE OF CHANCE Nano-vesicles from first trimester human placentae localized to the lungs, liver and kidneys 24 h after injection into pregnant mice (n = 7). Exposure of pregnant mice to placental nano-vesicles for 30 min in vivo increased the vasodilatory response of mesenteric arteries to acetylcholine, while exposure for 24 h had the opposite effect (P < 0.05, n = 6). These responses were prevented by L-NAME, an NO synthase inhibitor. Placental nano-vesicles did not affect the function of uterine arteries or mesenteric arteries from non-pregnant mice. Placental nano-vesicles rapidly interacted with endothelial cells via a combination of phagocytosis, endocytosis and cell surface binding in vitro. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION As it is not ethical to administer labelled placental nano-vesicles to pregnant women, pregnant CD1 mice were used as a model of pregnancy. WIDER IMPLICATIONS OF THE FINDINGS This is the first study to report the localization of placental nano-vesicles and their vascular effects in vivo. This work provides new insight into how the dramatic maternal cardiovascular adaptations to pregnancy may occur and indicates that placental extracellular vesicles may be important mediators of feto-maternal communication in a healthy pregnancy. STUDY FUNDING/COMPETING INTEREST(S) This research was supported by the Faculty of Medical and Health Science (FMHS) School of Medicine PBRF research fund to L.W.C. M.T. is a recipient of a University of Auckland Health Research Doctoral Scholarship and the Freemasons Postgraduate Scholarship. No authors have any competing interests to disclose.
Collapse
Affiliation(s)
- Mancy Tong
- Department of Obstetrics and Gynaecology, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| | - Joanna L Stanley
- Liggins Institute, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| | - Q Chen
- Department of Obstetrics and Gynaecology, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| | - Peter R Stone
- Department of Obstetrics and Gynaecology, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| | - Larry W Chamley
- Department of Obstetrics and Gynaecology, The University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand
| |
Collapse
|
147
|
Noguchi LM, Beigi RH. Treatment of infections during pregnancy: Progress and challenges. Birth Defects Res 2018; 109:387-390. [PMID: 28398676 DOI: 10.1002/bdr2.1005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/15/2016] [Accepted: 01/06/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND Emergence of Zika virus as a pathogen with important implications for perinatal outcomes highlights the need to identify safe and effective strategies for prevention and treatment of maternal infections. METHODS While substantial progress has been made in this area in recent years, significant regulatory and health systems barriers must still be overcome to identify and deliver evidence-based drug therapies for pregnant women. RESULTS We review progress and outstanding challenges associated with the identification and implementation of new treatment options for maternal infections. CONCLUSION We describe several strategies in use to optimize the application of existing evidence.Birth Defects Research 109:387-390, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lisa M Noguchi
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland
| | - Richard H Beigi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Hospital of UPMC, Pittsburgh, Pennsylvania
| |
Collapse
|
148
|
Mous DS, Kool HM, Burgisser PE, Buscop-van Kempen MJ, Nagata K, Boerema-de Munck A, van Rosmalen J, Dzyubachyk O, Wijnen RMH, Tibboel D, Rottier RJ. Treatment of rat congenital diaphragmatic hernia with sildenafil and NS-304, selexipag's active compound, at the pseudoglandular stage improves lung vasculature. Am J Physiol Lung Cell Mol Physiol 2018; 315:L276-L285. [PMID: 29745254 DOI: 10.1152/ajplung.00392.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Patients with congenital diaphragmatic hernia (CDH) often suffer from severe pulmonary hypertension, and the choice of current vasodilator therapy is mostly based on trial and error. Because pulmonary vascular abnormalities are already present early during development, we performed a study to modulate these pulmonary vascular changes at an early stage during gestation. Pregnant Sprague-Dawley rats were treated with nitrofen at day 9.5 of gestation (E9.5) to induce CDH in the offspring, and subsequently, the phosphodiesterase-5 inhibitor sildenafil and/or the novel prostaglandin-I receptor agonist selexipag (active compound NS-304) were administered from E17.5 until E20.5. The clinical relevant start of the treatment corresponds to week 20 of gestation in humans, when CDH is usually detected by ultrasound. CDH pups showed increased density of air saccules that was reverted after the use of only sildenafil. The pulmonary vascular wall was thickened, and right ventricular hypertrophy was present in the CDH group and improved both after single treatment with sildenafil or selexipag, whereas the combination therapy with both compounds did not have additive value. In conclusion, antenatal treatment with sildenafil improved airway morphogenesis and pulmonary vascular development, whereas selexipag only acted positively on pulmonary vascular development. The combination of both compounds did not act synergistically, probably because of a decreased efficiency of both compounds caused by cytochrome- P450 3A4 interaction and induction. These new insights create important possibilities for future treatment of pulmonary vascular abnormalities in CDH patients already in the antenatal period of life.
Collapse
Affiliation(s)
- Daphne S Mous
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Heleen M Kool
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Petra E Burgisser
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Marjon J Buscop-van Kempen
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Koji Nagata
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands.,Department of Pediatric Surgery, Reproductive and Developmental Medicine, Graduate School of Medical Sciences, Kyushu University , Fukuoka , Japan
| | - Anne Boerema-de Munck
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Joost van Rosmalen
- Department of Biostatistics, Erasmus Medical Center , Rotterdam , The Netherlands
| | - Oleh Dzyubachyk
- Department of Radiology, Leiden University Medical Center , Leiden , The Netherlands
| | - Rene M H Wijnen
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Dick Tibboel
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands
| | - Robbert J Rottier
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital , Rotterdam , The Netherlands.,Department of Cell Biology, Erasmus Medical Center , Rotterdam , The Netherlands
| |
Collapse
|
149
|
Pinto SR, Helal-Neto E, Paumgartten F, Felzenswalb I, Araujo-Lima CF, Martínez-Máñez R, Santos-Oliveira R. Cytotoxicity, genotoxicity, transplacental transfer and tissue disposition in pregnant rats mediated by nanoparticles: the case of magnetic core mesoporous silica nanoparticles. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:527-538. [PMID: 29688037 DOI: 10.1080/21691401.2018.1460603] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Whether in the cosmetic or as therapeutic, the use of nanoparticles has been increasing and taking on global proportion. However, there are few studies about the physical potential of long-term use or use in special conditions such as chronic, AIDS, pregnant women and other special health circumstances. In this context, the study of the mutagenicity and the transplacental passage represents an important and reliable model for the primary evaluation of potential health risks, especially maternal and child health. In this study we performed mutagenicity, cytotoxic and transplacental evaluation of magnetic core mesoporous silica nanoparticles, radiolabeled with 99mTc for determination of toxicogenic and embryonic/fetuses potential risk in animal model. Magnetic core mesoporous silica nanoparticles were produced and characterized by obtaining nanoparticles with a size of (58.9 ± 8.1 nm) in spherical shape and with intact magnetic core. The 99 m Tc radiolabeling process demonstrated high efficacy and stability in 98% yield over a period of 8 hours of stability. Mutagenicity assays were performed using Salmonella enteric serovar Typhimurium standard strains TA98, TA100 and TA102. Cytotoxicity assays were performed using WST-1. The transplacental evaluation assays were performed using the in vivo model with rats in two periods: embryonic and fetal stage. The results of both analyzes corroborate that the nanoparticles can i) generate DNA damage; ii) generate cytotoxic potential and iii) cross the transplantation barrier in both stages and bioaccumulates in both embryos and fetuses. The results suggest that complementary evaluations should be conducted in order to attest safety, efficacy and quality of nanoparticles before unrestricted approval of their use.
Collapse
Affiliation(s)
- Suyene Rocha Pinto
- a Nuclear Engineering Institute , Brazilian Nuclear Energy Commission , Rio de Janeiro , Brazil
| | - Edward Helal-Neto
- a Nuclear Engineering Institute , Brazilian Nuclear Energy Commission , Rio de Janeiro , Brazil
| | - Francisco Paumgartten
- b National School of Public Health , Oswaldo Cruz Foundation (FIOCRUZ) , Rio de Janeiro , Brazil
| | - Israel Felzenswalb
- c Departament of Biophysics and Biometrics, Environmental Mutagenesis Laboratory , Rio de Janeiro State University, Institute of Biology Roberto de Alcântara Gomes , Rio de Janeiro , Brazil
| | - Carlos Fernando Araujo-Lima
- c Departament of Biophysics and Biometrics, Environmental Mutagenesis Laboratory , Rio de Janeiro State University, Institute of Biology Roberto de Alcântara Gomes , Rio de Janeiro , Brazil
| | - Ramón Martínez-Máñez
- d Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) , Universitat Politècnica de València, Universitat de València , Valencia , Spain.,e Departamento de Química , Universidad Politécnica de Valencia , Valencia , Spain.,f CIBER de Bioingeniería , Biomateriales y Nanomedicina (CIBER-BBN) , Valencia , Spain
| | - Ralph Santos-Oliveira
- a Nuclear Engineering Institute , Brazilian Nuclear Energy Commission , Rio de Janeiro , Brazil.,g Laboratory of Nanoradiopharmaceuticals and Radiopharmacy , Zona Oeste State University , Rio de Janeiro , Brazil
| |
Collapse
|
150
|
Soo JY, Wiese MD, Berry MJ, Morrison JL. Does poor fetal growth influence the extent of fetal exposure to maternal medications? Pharmacol Res 2018; 130:74-84. [DOI: 10.1016/j.phrs.2018.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/18/2017] [Accepted: 02/01/2018] [Indexed: 10/18/2022]
|