1
|
Lemper M, Luetjens CM, Fuchs A, Hall P, Mathy FX, Long J, Cavagnaro J, Roy S, Thackaberry EA, Duda P. Subcutaneous zilucoplan: Evaluation of reproductive toxicology. Reprod Toxicol 2025; 134:108877. [PMID: 40049247 DOI: 10.1016/j.reprotox.2025.108877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
To further understand the safety profile of zilucoplan, reproductive toxicology studies in non-human primates (NHPs) were performed, including a male fertility study and a combined embryo-foetal development (EFD) and enhanced pre- and postnatal development (ePPND) study. Human transplacental transfer of zilucoplan was examined in an ex vivo human placental perfusion model of foetal exposure during pregnancy. By comparison with the positive control, a low transfer rate of 0.5 % was observed with a target blood zilucoplan concentration that mimics the therapeutic dose of 0.3 mg/kg used in clinical trials in patients with generalised myasthenia gravis. In each in vivo study, daily subcutaneous zilucoplan 1.0, 2.0 or 4.0 mg/kg or vehicle control were randomly assigned. Six male cynomolgus monkeys/group received treatment for 13 weeks (male fertility), 4 females/group received treatment for 80 days (EFD) and 16 females/group received treatment from gestation day 20 to delivery (∼140 days, ePPND). Developmental, reproductive and toxicokinetic effects were analysed. No zilucoplan-related reproductive effects were observed in the male fertility study. There were no effects on pregnancy outcome, number of viable foetuses or foetal development in the combined EFD/ePPND study. Overall pregnancy loss (including stillbirths and death during birth) was 25.0 % (4/16) in the control group compared with 37.5 % (6/16) in the zilucoplan-treated groups and was within the published range of pregnancy loss in cynomolgus monkeys. These reproductive toxicity studies demonstrate no adverse effect of zilucoplan on male fertility or maternal or embryo-foetal outcomes, and no pre- or postnatal toxicity in NHPs receiving daily zilucoplan. Data from the ex vivo placental transfer model and the lack of effect of FcRn-mediated transfer on zilucoplan suggest that placental transfer is low.
Collapse
|
2
|
Kuzniewicz MW, Escobar GJ, Forquer H, Li S, Shu D, Kipnis P, Fischer A, Puopolo KM. Update to the Neonatal Early-Onset Sepsis Calculator Utilizing a Contemporary Cohort. Pediatrics 2024; 154:e2023065267. [PMID: 39314183 DOI: 10.1542/peds.2023-065267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The Kaiser Permanente Neonatal Early-Onset Sepsis (EOS) Calculator has been an effective tool for risk stratification to safely reduce newborn antibiotic exposure. The calculator was derived from data on infants born between 1993 and 2007. Since that time, US obstetric practice has adopted universal antepartum screening for group B Streptococcus and intrapartum antibiotic prophylaxis guidance has changed. Our objective was to update the EOS calculator using a contemporary birth cohort and determine the effect of these changes on EOS case ascertainment and antibiotic recommendations. METHODS The study included infants born at ≥35 weeks' gestation at 14 hospitals between January 2010 and December 2020 (n = 412 595 infants, EOS cases = 113). Model coefficients were re-estimated and the point estimates of the likelihood ratios for clinical status used to calculate the posterior probability of EOS. We compared the number of EOS cases correctly identified by each model (sensitivity) and the proportion of infants for whom empirical antibiotics are recommended. RESULTS The original model had a sensitivity of 0.76 (95% confidence interval 0.63-0.85), while the updated model had a sensitivity of 0.80 (95% confidence interval 0.68-0.89), P = .15. The recommended empirical antibiotic use was 3.5% with the original model and 3.7% with the updated model, P < .0001. For each additional case identified by the updated model, an additional 158 infants would be treated with antibiotics. CONCLUSIONS Both the original and updated EOS calculators are effective tools for quantifying EOS risk among infants born at ≥35 weeks' gestation.
Collapse
Affiliation(s)
- Michael W Kuzniewicz
- Division of Research
- Department of Pediatrics, Kaiser Permanente Northern California, Oakland, California
- Division of Neonatology, University of California, San Francisco, San Francisco, California
| | | | | | | | - Di Shu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
- Clinical Futures
| | | | - Allen Fischer
- Department of Pediatrics, Kaiser Permanente Northern California, Oakland, California
| | - Karen M Puopolo
- Clinical Futures
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Wang X, Patrikeeva S, Nanovskaya T, Bryant V. Development and validation of HPLC-ultraviolet method for quantitative determination of pritelivir in human placental perfusion medium. Biomed Chromatogr 2024; 38:e5888. [PMID: 38727008 PMCID: PMC11262560 DOI: 10.1002/bmc.5888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 06/20/2024]
Abstract
A simple and reliable HPLC-ultraviolet (HPLC-UV) method was developed and validated for the quantification of pritelivir in the samples of medium from the experiments utilizing the ex vivo technique of dual perfusion of the human placental lobule. Phenacetin was used as an internal standard (IS) in our HPLC-UV method. Chromatographic separation of pritelivir and phenacetin was achieved on a Waters Symmetry C18 HPLC column (100 × 2.1 mm, 3.5 μm) at ambient temperature (22-25°C). The mobile phase was composed of 50% methanol in deionized water (v/v), the flow rate for isocratic elution was established at 0.25 mL/min, and the detection wavelength for pritelivir and IS was set at 254 nm. Pritelivir and IS were extracted with the protein precipitation method using methanol as a solvent. The calibration curve for pritelivir exhibited linearity (r2 > 0.99) within the concentration range from 0.155 to 6.62 μg/mL. Within- and between-day accuracy ranged from 97% to 110% with relative standard deviation (RSD) values not exceeding 10%. The extraction recovery of pritelivir and IS ranged from 89% to 91% with RSD not exceeding 7%. Pritelivir was stable under the storage and sample handling conditions. This validated HPLC-UV method was utilized to quantify pritelivir in the placental perfusion medium samples, and the resulting concentrations were authenticated with incurred sample reanalysis to confirm the reliability of the method.
Collapse
Affiliation(s)
- Xiaoming Wang
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Svetlana Patrikeeva
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Tatiana Nanovskaya
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Valentina Bryant
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
4
|
Miao S, Yin J, Liu S, Zhu Q, Liao C, Jiang G. Maternal-Fetal Exposure to Antibiotics: Levels, Mother-to-Child Transmission, and Potential Health Risks. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:8117-8134. [PMID: 38701366 DOI: 10.1021/acs.est.4c02018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Due to its widespread applications in various fields, antibiotics are continuously released into the environment and ultimately enter the human body through diverse routes. Meanwhile, the unreasonable use of antibiotics can also lead to a series of adverse outcomes. Pregnant women and developing fetuses are more susceptible to the influence of external chemicals than adults. The evaluation of antibiotic exposure levels through questionnaire surveys or prescriptions in medical records and biomonitoring-based data shows that antibiotics are frequently prescribed and used by pregnant women around the world. Antibiotics may be transmitted from mothers to their offspring through different pathways, which then adversely affect the health of offspring. However, there has been no comprehensive review on antibiotic exposure and mother-to-child transmission in pregnant women so far. Herein, we summarized the exposure levels of antibiotics in pregnant women and fetuses, the exposure routes of antibiotics to pregnant women, and related influencing factors. In addition, we scrutinized the potential mechanisms and factors influencing the transfer of antibiotics from mother to fetus through placental transmission, and explored the adverse effects of maternal antibiotic exposure on fetal growth and development, neonatal gut microbiota, and subsequent childhood health. Given the widespread use of antibiotics and the health threats posed by their exposure, it is necessary to comprehensively track antibiotics in pregnant women and fetuses in the future, and more in-depth biological studies are needed to reveal and verify the mechanisms of mother-to-child transmission, which is crucial for accurately quantifying and evaluating fetal health status.
Collapse
Affiliation(s)
- Shiyu Miao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jia Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingqing Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunyang Liao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Yan Y, Wang Q, Wu W, Yi H, Xie F. Evaluation of Various Approaches to Estimate Transplacental Clearance of Vancomycin for Predicting Fetal Concentrations using a Maternal-Fetal Physiologically Based Pharmacokinetic Model. Pharm Res 2024; 41:899-910. [PMID: 38684563 DOI: 10.1007/s11095-024-03705-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Evaluating drug transplacental clearance is vital for forecasting fetal drug exposure. Ex vivo human placenta perfusion experiments are the most suitable approach for this assessment. Various in silico methods are also proposed. This study aims to compare these prediction methods for drug transplacental clearance, focusing on the large molecular weight drug vancomycin (1449.3 g/mol), using maternal-fetal physiologically based pharmacokinetic (m-f PBPK) modeling. METHODS Ex vivo human placenta perfusion experiments, in silico approaches using intestinal permeability as a substitute (quantitative structure property relationship (QSPR) model and Caco-2 permeability in vitro-in vivo correlation model) and midazolam calibration model with Caco-2 scaling were assessed for determining the transplacental clearance (CLPD) of vancomycin. The m-f PBPK model was developed stepwise using Simcyp, incorporating the determined CLPD values as a crucial input parameter for transplacental kinetics. RESULTS The developed PBPK model of vancomycin for non-pregnant adults demonstrated excellent predictive performance. By incorporating the CLPD parameterization derived from ex vivo human placenta perfusion experiments, the extrapolated m-f PBPK model consistently predicted maternal and fetal concentrations of vancomycin across diverse doses and distinct gestational ages. However, when the CLPD parameter was derived from alternative prediction methods, none of the extrapolated maternal-fetal PBPK models produced fetal predictions in line with the observed data. CONCLUSION Our study showcased that combination of ex vivo human placenta perfusion experiments and m-f PBPK model has the capability to predict fetal exposure for the large molecular weight drug vancomycin, whereas other in silico approaches failed to achieve the same level of accuracy.
Collapse
Affiliation(s)
- Yunan Yan
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China
| | - Qiushi Wang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China
| | - Wei Wu
- The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Hanxi Yi
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo Road 172, Changsha, 410013, China.
| |
Collapse
|
6
|
Coggins SA, Puopolo KM. Neonatal Group B Streptococcus Disease. Pediatr Rev 2024; 45:63-73. [PMID: 38296778 PMCID: PMC10919294 DOI: 10.1542/pir.2023-006154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
Group B Streptococcus (GBS) is an important cause of neonatal sepsis in term and preterm infants. Because GBS colonizes human genitourinary and gastrointestinal tracts, a significant focus of neonatal GBS disease prevention is to interrupt vertical transmission of GBS from mother to infant during parturition. Routine antepartum GBS screening in pregnant women, as well as widespread use of intrapartum antibiotic prophylaxis, have aided in overall reductions in neonatal GBS disease during the past 3 decades. However, neonatal GBS disease persists and may cause mortality and significant short- and long-term morbidity among survivors. Herein, we highlight contemporary epidemiology, microbial pathogenesis, and the clinical presentation spectrum associated with neonatal GBS disease. We summarize obstetric recommendations for antenatal GBS screening, indications for intrapartum antibiotic prophylaxis, and considerations for antibiotic selection. Finally, we review national guidelines for risk assessment and management of infants at risk for GBS disease.
Collapse
MESH Headings
- Infant
- Pregnancy
- Female
- Infant, Newborn
- Humans
- Pregnancy Complications, Infectious/diagnosis
- Pregnancy Complications, Infectious/drug therapy
- Pregnancy Complications, Infectious/epidemiology
- Streptococcal Infections/diagnosis
- Streptococcal Infections/drug therapy
- Streptococcal Infections/epidemiology
- Infant, Premature
- Anti-Bacterial Agents/therapeutic use
- Infant, Newborn, Diseases/diagnosis
- Infant, Newborn, Diseases/therapy
- Streptococcus agalactiae
- Infectious Disease Transmission, Vertical/prevention & control
Collapse
Affiliation(s)
- Sarah A. Coggins
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, US
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Karen M. Puopolo
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, US
- Department of Pediatrics, University of Pennsylvania, Philadelphia, PA, USA
- Clinical Futures, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
7
|
Calis P, Vojtech L, Hladik F, Gravett MG. A review of ex vivo placental perfusion models: an underutilized but promising method to study maternal-fetal interactions. J Matern Fetal Neonatal Med 2022; 35:8823-8835. [PMID: 34818981 PMCID: PMC9126998 DOI: 10.1080/14767058.2021.2005565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 02/08/2023]
Abstract
Studying the placenta can provide information about the mechanistic pathways of pregnancy disease. However, analyzing placental tissues and manipulating placental function in real-time during pregnancy is not feasible. The ex vivo placental perfusion model allows observing important aspects of the physiology and pathology of the placenta, while maintaining its viability and functional integrity, and without causing harm to mother or fetus. In this review, we describe and compare setups for this technically complex model and summarize outcomes from various published studies. We hope that our review will encourage wider use of ex vivo placental perfusion, which in turn would generate more knowledge to improve pregnancy outcomes.
Collapse
Affiliation(s)
- Pinar Calis
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Michael G. Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| |
Collapse
|
8
|
van Hove H, Mathiesen L, Freriksen J, Vähäkangas K, Colbers A, Brownbill P, Greupink R. Placental transfer and vascular effects of pharmaceutical drugs in the human placenta ex vivo: A review. Placenta 2022; 122:29-45. [DOI: 10.1016/j.placenta.2022.03.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
9
|
Ren Z, Bremer AA, Pawlyk AC. Drug development research in pregnant and lactating women. Am J Obstet Gynecol 2021; 225:33-42. [PMID: 33887238 DOI: 10.1016/j.ajog.2021.04.227] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022]
Abstract
Pregnant and lactating women are considered "therapeutic orphans" because they generally have been excluded from clinical drug research and the drug development process owing to legal, ethical, and safety concerns. Most medications prescribed for pregnant and lactating women are used "off-label" because most of the clinical approved medications do not have appropriate drug labeling information for pregnant and lactating women. Medications that lack human safety data on use during pregnancy and lactation may pose potential risks for adverse effects in pregnant and lactating women as well as risks of teratogenic effects to their unborn and newborn babies. Federal policy requiring the inclusion of women in clinical research and trials led to considerable changes in research design and practice. Despite more women being included in clinical research and trials, the inclusion of pregnant and lactating women in drug research and clinical trials remains limited. A recent revision to the "Common Rule" that removed pregnant women from the classification as a "vulnerable" population may change the culture of drug research and drug development in pregnant and lactating women. This review article provides an overview of medications studied by the Obstetric-Fetal Pharmacology Research Units Network and Centers and describes the challenges in current obstetrical pharmacology research and alternative strategies for future research in precision therapeutics in pregnant and lactating women. Implementation of the recommendations of the Task Force on Research Specific to Pregnant Women and Lactating Women can provide legislative requirements and opportunities for research focused on pregnant and lactating women.
Collapse
Affiliation(s)
- Zhaoxia Ren
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD.
| | - Andrew A Bremer
- Pediatric Growth and Nutrition Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD; Pregnancy and Perinatology Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| | - Aaron C Pawlyk
- Obstetric and Pediatric Pharmacology and Therapeutics Branch, the Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD
| |
Collapse
|
10
|
Nadeau HCG, Edwards RK. Prophylaxis Against Early-onset Group B Streptococcus Infections in Pregnant Women Who Are Allergic to Penicillin. Clin Obstet Gynecol 2020; 62:771-780. [PMID: 30998602 DOI: 10.1097/grf.0000000000000455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Group B Streptococcus (GBS) infection remains a significant cause of neonatal morbidity and mortality. Adoption of screening for maternal genital tract colonization and intrapartum antibiotic prophylaxis has significantly reduced early-onset neonatal GBS infections. For women with an allergy to penicillin, recommended agents for prophylaxis have been well-outlined, but compliance with guideline recommendations is poor. There have been ongoing efforts in vaccine development, but no vaccination currently is available for either preconception or antenatal administration. This article will review established screening techniques, intrapartum antibiotic prophylactic regimens, and management specifically of the penicillin-allergic pregnant woman who is colonized with GBS.
Collapse
Affiliation(s)
- Hugh C G Nadeau
- Section of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | | |
Collapse
|
11
|
|
12
|
Evaluation of Fetal and Maternal Vancomycin-Induced Kidney Injury during Pregnancy in a Rat Model. Antimicrob Agents Chemother 2019; 63:AAC.00761-19. [PMID: 31332061 DOI: 10.1128/aac.00761-19] [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] [Received: 04/08/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Previous literature suggests that maternal vancomycin crosses the placental barrier to the fetus. Further, early animal studies indicated that kidney injury was not observed in the progeny. These studies were conducted prior to the availability of sensitive biomarkers for kidney injury. Therefore, a previous finding of no renal damage to the infant may be misleading. Vancomycin was administered intravenously to pregnant rats at a dose of 250 mg/kg of body weight/day (N = 6 per trimester) on three consecutive gestational days (GD) during trimesters 1, 2, and 3 (T1, T2, and T3, respectively) in three independent cohorts. The dams carried to term and delivered vaginally on GD 21. Kidneys were harvested from dams and pups and homogenized. Samples were prepared by protein precipitation and injected in a liquid chromatography tandem mass spectrometer, and vancomycin was quantified. The kidney tissue homogenate from dams and pups were analyzed for kidney injury molecule-1 (KIM-1). As trimesters progressed, the quantity of vancomycin increased linearly in the kidneys of both rat dams and pups (P < 0.0001 for T1 and T3, P < 0.0001 for T2 and T3, and P < 0.0001 for T3 and T3 control for both rat dams and pups). KIM-1 concentrations in pup kidneys were significantly higher when dams were administered vancomycin in trimesters 1 (P = 0.0001) and 2 (P = 0.0024) than in controls in trimester 3. Data demonstrate persistence of vancomycin in maternal and rat pup kidneys in all three trimesters of pregnancy with associated damage to the kidney, as indicated by expression of KIM-1.
Collapse
|
13
|
Viel-Theriault I, Fell DB, Grynspan D, Redpath S, Thampi N. The transplacental passage of commonly used intrapartum antibiotics and its impact on the newborn management: A narrative review. Early Hum Dev 2019; 135:6-10. [PMID: 31177037 DOI: 10.1016/j.earlhumdev.2019.05.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 11/30/2022]
Abstract
Neonates exposed to intra-amniotic infection are at increased risk of early-onset sepsis. Administration of antibiotics to the mother may offer some protection, however a comprehensive description of the determinants influencing their transplacental passage and delivery to the fetus has not been performed. While penicillin G, ampicillin, cefazolin and gentamicin reach therapeutic levels in the fetal serum rapidly following maternal administration, the transfer of second-line intrapartum antimicrobials, such as vancomycin and clindamycin, is slower and less predictable. Erythromycin, used in the context of preterm premature rupture of the membranes, has suboptimal influx into the fetal compartment. This evidence is predominantly drawn from term pregnancies and situations of low infectious risk; however, prematurity may negatively influence fetal exposure to intrapartum antibiotics. Optimal fetal antimicrobial concentrations to target are poorly defined and the extent to which our review findings apply to preterm early-onset neonatal sepsis prevention is unclear. Interpretation of blood cultures drawn in neonates with expected circulating levels of maternal antimicrobials above the minimal inhibitory concentration for Group B Streptococcus is challenging despite the use of contemporary optimized blood culture media.
Collapse
Affiliation(s)
- I Viel-Theriault
- Division of Infectious Diseases, Department of Pediatrics, The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada.
| | - D B Fell
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - D Grynspan
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - S Redpath
- Division of Neonatology, Department of Pediatrics, The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - N Thampi
- Division of Infectious Diseases, Department of Pediatrics, The Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| |
Collapse
|
14
|
Puopolo KM, Lynfield R, Cummings JJ, Hand I, Adams-Chapman I, Poindexter B, Stewart DL, Aucott SW, Goldsmith JP, Mowitz M, Watterberg K, Maldonado YA, Zaoutis TE, Banerjee R, Barnett ED, Campbell JD, Gerber JS, Kourtis AP, Munoz FM, Nolt D, Nyquist AC, O’Leary ST, Sawyer MH, Steinbach WJ, Zangwill K, COMMITTEE ON FETUS AND NEWBORN, COMMITTEE ON INFECTIOUS DISEASES. Management of Infants at Risk for Group B Streptococcal Disease. Pediatrics 2019; 144:peds.2019-1881. [PMID: 31285392 DOI: 10.1542/peds.2019-1881] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Group B streptococcal (GBS) infection remains the most common cause of neonatal early-onset sepsis and a significant cause of late-onset sepsis among young infants. Administration of intrapartum antibiotic prophylaxis is the only currently available effective strategy for the prevention of perinatal GBS early-onset disease, and there is no effective approach for the prevention of late-onset disease. The American Academy of Pediatrics joins with the American College of Obstetricians and Gynecologists to reaffirm the use of universal antenatal microbiologic-based testing for the detection of maternal GBS colonization to facilitate appropriate administration of intrapartum antibiotic prophylaxis. The purpose of this clinical report is to provide neonatal clinicians with updated information regarding the epidemiology of GBS disease as well current recommendations for the evaluation of newborn infants at risk for GBS disease and for treatment of those with confirmed GBS infection. This clinical report is endorsed by the American College of Obstetricians and Gynecologists (ACOG), July 2019, and should be construed as ACOG clinical guidance.
Collapse
Affiliation(s)
- Karen M. Puopolo
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Ruth Lynfield
- Minnesota Department of Health, St Paul, Minnesota; and
| | - James J. Cummings
- Departments of Pediatrics and Bioethics, Alden March Bioethics Institute, Albany Medical College, Albany, New York
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Prevention of Group B Streptococcal Early-Onset Disease in Newborns: ACOG Committee Opinion, Number 782. Obstet Gynecol 2019; 134:1. [PMID: 31241599 DOI: 10.1097/aog.0000000000003334] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Group B streptococcus (GBS) is the leading cause of newborn infection. The primary risk factor for neonatal GBS early-onset disease (EOD) is maternal colonization of the genitourinary and gastrointestinal tracts. Approximately 50% of women who are colonized with GBS will transmit the bacteria to their newborns. Vertical transmission usually occurs during labor or after rupture of membranes. In the absence of intrapartum antibiotic prophylaxis, 1-2% of those newborns will develop GBS EOD. Other risk factors include gestational age of less than 37 weeks, very low birth weight, prolonged rupture of membranes, intraamniotic infection, young maternal age, and maternal black race. The key obstetric measures necessary for effective prevention of GBS EOD continue to include universal prenatal screening by vaginal-rectal culture, correct specimen collection and processing, appropriate implementation of intrapartum antibiotic prophylaxis, and coordination with pediatric care providers. The American College of Obstetricians and Gynecologists now recommends performing universal GBS screening between 36 0/7 and 37 6/7 weeks of gestation. All women whose vaginal-rectal cultures at 36 0/7-37 6/7 weeks of gestation are positive for GBS should receive appropriate intrapartum antibiotic prophylaxis unless a prelabor cesarean birth is performed in the setting of intact membranes. Although a shorter duration of recommended intrapartum antibiotics is less effective than 4 or more hours of prophylaxis, 2 hours of antibiotic exposure has been shown to reduce GBS vaginal colony counts and decrease the frequency of a clinical neonatal sepsis diagnosis. Obstetric interventions, when necessary, should not be delayed solely to provide 4 hours of antibiotic administration before birth. This Committee Opinion, including , , and , updates and replaces the obstetric components of the CDC 2010 guidelines, "Prevention of Perinatal Group B Streptococcal Disease: Revised Guidelines From CDC, 2010."
Collapse
|
16
|
Abstract
Antimicrobial medications are the most commonly used medications in the neonatal intensive care unit. Antibiotics are used for infection prophylaxis, empiric treatment, and definitive treatment of confirmed infection. The choice of medication should be informed by the epidemiology and microbiology of infection in specific clinical scenarios and by the clinical condition of the infant. Understanding evolving pathogen susceptibility to antimicrobials and key pharmacotherapy determinants in neonates can inform optimal antibiotic use.
Collapse
Affiliation(s)
- Sagori Mukhopadhyay
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA
| | - Kelly C Wade
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA
| | - Karen M Puopolo
- Section on Newborn Medicine, Pennsylvania Hospital, Philadelphia, PA, USA; Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; CHOP Newborn Care, Pennsylvania Hospital, 800 Spruce Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
17
|
Roy S, Nanovskaya T, Patrikeeva S, Cochran E, Parge V, Guess J, Schaeck J, Choudhury A, Ahmed M, Ling LE. M281, an anti-FcRn antibody, inhibits IgG transfer in a human ex vivo placental perfusion model. Am J Obstet Gynecol 2019; 220:498.e1-498.e9. [PMID: 30849355 DOI: 10.1016/j.ajog.2019.02.058] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 02/22/2019] [Accepted: 02/27/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND The transfer of pathogenic immunoglobulin G antibodies from mother to fetus is a critical step in the pathophysiology of alloimmune and autoimmune diseases of the fetus and neonate. Immunoglobulin G transfer across the human placenta to the fetus is mediated by the neonatal Fc receptor, and blockade of the neonatal Fc receptor may provide a therapeutic strategy to prevent or minimize pathological events associated with immune-mediated diseases of pregnancy. M281 is a fully human, aglycosylated monoclonal immunoglobulin G1 antineonatal Fc receptor antibody that has been shown to block the neonatal Fc receptor with high affinity in nonclinical studies and in a phase 1 study in healthy volunteers. OBJECTIVE The objective of the study was to determine the transplacental transfer of M281 and its potential to inhibit transfer of immunoglobulin G from maternal to fetal circulation. STUDY DESIGN To determine the concentration of M281 required for rapid cellular uptake and complete saturation of the neonatal Fc receptor in placental trophoblasts, primary human villous trophoblasts were incubated with various concentrations of M281 in a receptor occupancy assay. The placental transfer of M281, immunoglobulin G, and immunoglobulin G in the presence of M281 was studied using the dually perfused human placental lobule model. Immunoglobulin G transfer was established using a representative immunoglobulin G molecule, adalimumab, a human immunoglobulin G1 monoclonal antibody, at a concentration of 270 μg/mL. Inhibition of immunoglobulin G transfer by M281 was determined by cotransfusing 270 μg/mL of adalimumab with 10 μg/mL or 300 μg/mL of M281. Concentrations of adalimumab and M281 in sample aliquots from maternal and fetal circuits were analyzed using a sandwich enzyme-linked immunosorbent assay and Meso Scale Discovery assay, respectively. RESULTS In primary human villous trophoblasts, the saturation of the neonatal Fc receptor by M281 was observed within 30-60 minutes at 0.15-5.0 μg/mL, suggesting rapid blockade of neonatal Fc receptor in placental cells. The transfer rate of adalimumab (0.23% ± 0.21%) across dually perfused human placental lobule was significantly decreased by 10 μg/mL and 300 μg/mL of M281 to 0.07 ± 0.01% and 0.06 ± 0.01%, respectively. Furthermore, the transfer rate of M281 was 0.002% ± 0.02%, approximately 100-fold lower than that of adalimumab. CONCLUSION The significant inhibition of immunoglobulin G transfer across the human placental lobule by M281 and the minimal transfer of M281 supports the development of M281 as a novel agent for the treatment of fetal and neonatal diseases caused by transplacental transfer of alloimmune and autoimmune pathogenic immunoglobulin G antibodies.
Collapse
|
18
|
Aengenheister L, Keevend K, Muoth C, Schönenberger R, Diener L, Wick P, Buerki-Thurnherr T. An advanced human in vitro co-culture model for translocation studies across the placental barrier. Sci Rep 2018; 8:5388. [PMID: 29599470 PMCID: PMC5876397 DOI: 10.1038/s41598-018-23410-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/12/2018] [Indexed: 01/10/2023] Open
Abstract
Although various drugs, environmental pollutants and nanoparticles (NP) can cross the human placental barrier and may harm the developing fetus, knowledge on predictive placental transfer rates and the underlying transport pathways is mostly lacking. Current available in vitro placental transfer models are often inappropriate for translocation studies of macromolecules or NPs and do not consider barrier function of placental endothelial cells (EC). Therefore, we developed a human placental in vitro co-culture transfer model with tight layers of trophoblasts (BeWo b30) and placental microvascular ECs (HPEC-A2) on a low-absorbing, 3 µm porous membrane. Translocation studies with four model substances and two polystyrene (PS) NPs across the individual and co-culture layers revealed that for most of these compounds, the trophoblast and the EC layer both demonstrate similar, but not additive, retention capacity. Only the paracellular marker Na-F was substantially more retained by the BeWo layer. Furthermore, simple shaking, which is often applied to mimic placental perfusion, did not alter translocation kinetics compared to static exposure. In conclusion, we developed a novel placental co-culture model, which provides predictive values for translocation of a broad variety of molecules and NPs and enables valuable mechanistic investigations on cell type-specific placental barrier function.
Collapse
Affiliation(s)
- Leonie Aengenheister
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Kerda Keevend
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Carina Muoth
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - René Schönenberger
- UTOX, EAWAG, Swiss Federal Institute of Aquatic Science and Technology, Ueberlandstrasse 133, 8600, Dübendorf, Switzerland
| | - Liliane Diener
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Peter Wick
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland
| | - Tina Buerki-Thurnherr
- Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, Lerchenfeldstrasse 5, 9014, St. Gallen, Switzerland.
| |
Collapse
|
19
|
Brownbill P, Sebire N, McGillick EV, Ellery S, Murthi P. Ex Vivo Dual Perfusion of the Human Placenta: Disease Simulation, Therapeutic Pharmacokinetics and Analysis of Off-Target Effects. Methods Mol Biol 2018; 1710:173-189. [PMID: 29197003 DOI: 10.1007/978-1-4939-7498-6_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In recent years ex vivo dual perfusion of the human placental lobule is seeing an international renaissance in its application to understanding fetal health and development. Here, we discuss the methods and uses of this technique in the evaluation of (1) vascular function, (2) transplacental clearance, (3) hemodynamic and oxygenation changes associated with pregnancy complications on placental structure and function, and (4) placental toxicology and post-perfusion evaluation of tissue architecture.
Collapse
Affiliation(s)
- Paul Brownbill
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
- St. Mary's Hospital, Central Manchester University Hospitals, NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Neil Sebire
- Institute of Child Health, University College London, London, UK
| | - Erin V McGillick
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- The Department of Obstetrics and Gynecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Stacey Ellery
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- The Department of Obstetrics and Gynecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Padma Murthi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- The Department of Obstetrics and Gynecology, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
20
|
Affiliation(s)
- Craig V. Towers
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Tennessee Medical Center, Knoxville, TN, USA
| | - Beth Weitz
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Tennessee Medical Center, Knoxville, TN, USA
| |
Collapse
|
21
|
Das B, Sarkar C, Das D, Gupta A, Kalra A, Sahni S. Telavancin: a novel semisynthetic lipoglycopeptide agent to counter the challenge of resistant Gram-positive pathogens. Ther Adv Infect Dis 2017; 4:49-73. [PMID: 28634536 PMCID: PMC5467880 DOI: 10.1177/2049936117690501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Telavancin (TD-6424), a semisynthetic lipoglycopeptide vancomycin-derivative, is a novel antimicrobial agent developed by Theravance for overcoming resistant Gram-positive bacterial infections, specifically methicillin-resistant Staphylococcus aureus (MRSA). The US Food and Drug Administration (USFDA) had approved telavancin in 2009 for the treatment of complicated skin and skin structure infections (cSSSIs) caused by Gram-positive bacteria, including MRSA (S. aureus, Streptococcus agalactiae, Streptococcus pyogenes, Streptococcus anginosus group, or Enterococcus faecalis). Telavancin has two proposed mechanisms of action. In vitro, telavancin has a rapid, concentration-dependent bactericidal effect, due to disruption of cell membrane integrity. Telavancin has demonstrable in vitro activity against aerobic and anaerobic Gram-positive bacteria. Telavancin and vancomycin have similar spectra of activity. Gram-negative bacteria are usually non-susceptible to telavancin. Telavancin has been successfully tested in various animal models of bacteremia, endocarditis, meningitis, and pneumonia. Phase II Telavancin versus Standard Therapy for Treatment of Complicated Skin and Soft-Tissue Infections due to Gram-Positive Bacteria (FAST 1 and FAST 2) and phase III [Assessment of Telavancin in Complicated Skin and Skin Structure Infections 1 (ATLAS 1 and ATLAS 2)] clinical trials have been conducted for evaluating telavancin's efficacy and safety in cSSSIs. Phase III clinical trials have been carried out for evaluating telavancin's safety and efficacy in nosocomial pneumonia [Assessment of Telavancin for Treatment of Hospital acquired Pneumonia 1 and 2 (ATTAIN 1 and ATTAIN 2)]. A phase II randomized, double-blind, clinical trial has been carried out for evaluating telavancin's safety and efficacy in uncomplicated S. aureus bacteremia [Telavancin for Treatment of Uncomplicated S. aureus Bacteremia (ASSURE)]. Pacemaker lead-related infective endocarditis due to a vancomycin intermediate S. aureus (VISA) strain (non-daptomycin susceptible) was successfully treated with parenteral telavancin for 8 weeks. Telavancin extensively binds to serum albumin (~93%) and has a relatively small volume of distribution. Telavancin is not biotransformed by any cytochrome P450 microsomal enzymes and excreted mainly in the urine. Though well-tolerated, worrisome adverse effects, including renal dysfunction and QTc prolongation are of potential concern. Given its extensive binding to plasma proteins, long half-life, and a long post-antibiotic effect, it represents a promising addition to the therapeutic armamentarium in combating infections caused by resistant Gram-positive pathogens, namely, MRSA.
Collapse
Affiliation(s)
- Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Chayna Sarkar
- Department of Pharmacology & Clinical Pharmacology, North Eastern Indira Gandhi Regional Institute of Health & Medical Sciences (NEIGRIHMS) Shillong, Shillong, India
| | - Debasmita Das
- Department of Computer Science & Engineering, Faculty of Engineering, Manipal University Jaipur, Dehmi Kalan, Jaipur Ajmer Expressway, Rajasthan, India
| | - Amit Gupta
- Department of Surgery, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Arnav Kalra
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| | - Shubham Sahni
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh, India
| |
Collapse
|
22
|
Grafmueller S, Manser P, Diener L, Diener PA, Maeder-Althaus X, Maurizi L, Jochum W, Krug HF, Buerki-Thurnherr T, von Mandach U, Wick P. Bidirectional Transfer Study of Polystyrene Nanoparticles across the Placental Barrier in an ex Vivo Human Placental Perfusion Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:1280-6. [PMID: 25956008 PMCID: PMC4671239 DOI: 10.1289/ehp.1409271] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 05/05/2015] [Indexed: 05/08/2023]
Abstract
BACKGROUND Nanoparticle exposure in utero might not be a major concern yet, but it could become more important with the increasing application of nanomaterials in consumer and medical products. Several epidemiologic and in vitro studies have shown that nanoparticles can have potential toxic effects. However, nanoparticles also offer the opportunity to develop new therapeutic strategies to treat specifically either the pregnant mother or the fetus. Previous studies mainly addressed whether nanoparticles are able to cross the placental barrier. However, the transport mechanisms underlying nanoparticle translocation across the placenta are still unknown. OBJECTIVES In this study we examined which transport mechanisms underlie the placental transfer of nanoparticles. METHODS We used the ex vivo human placental perfusion model to analyze the bidirectional transfer of plain and carboxylate modified polystyrene particles in a size range between 50 and 300 nm. RESULTS We observed that the transport of polystyrene particles in the fetal to maternal direction was significantly higher than for the maternal to fetal direction. Regardless of their ability to cross the placental barrier and the direction of perfusion, all polystyrene particles accumulated in the syncytiotrophoblast of the placental tissue. CONCLUSIONS Our results indicate that the syncytiotrophoblast is the key player in regulating nanoparticle transport across the human placenta. The main mechanism underlying this translocation is not based on passive diffusion, but is likely to involve an active, energy-dependent transport pathway. These findings will be important for reproductive toxicology as well as for pharmaceutical engineering of new drug carriers.
Collapse
|
23
|
Padberg S. Anti-infective Agents. DRUGS DURING PREGNANCY AND LACTATION 2015. [PMCID: PMC7150338 DOI: 10.1016/b978-0-12-408078-2.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
24
|
Abstract
Pregnancy is associated with a variety of physiological changes that can alter the pharmacokinetics and pharmacodynamics of several drugs. However, limited data exists on the pharmacokinetics and pharmacodynamics of the majority of the medications used in pregnancy. In this article, we first describe basic concepts (drug absorption, bioavailability, distribution, metabolism, elimination, and transport) in pharmacokinetics. Then, we discuss several physiological changes that occur during pregnancy that theoretically affect absorption, distribution, metabolism, and elimination. Further, we provide a brief review of the literature on the clinical pharmacokinetic studies performed in pregnant women in recent years. In general, pregnancy increases the clearance of several drugs and correspondingly decreases drug exposure during pregnancy. Based on current drug exposure measurements during pregnancy, alterations in the dose or dosing regimen of certain drugs are essential during pregnancy. More pharmacological studies in pregnant women are needed to optimize drug therapy in pregnancy.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 718 Salk Hall, 3501 Terrace St, Pittsburgh, PA 15261
| | - Mary F. Hebert
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA,Department of Obstetrics and Gynecology, School of Medicine University of Washington, Seattle, WA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 718 Salk Hall, 3501 Terrace St, Pittsburgh, PA 15261; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA; Thomas Starzl Transplantation Institute, Pittsburgh, PA; McGovern Institute for Regenerative Medicine, Pittsburgh, PA; Magee Womens Research Institute, Pittsburgh, PA.
| |
Collapse
|
25
|
Onwuchuruba CN, Towers CV, Howard BC, Hennessy MD, Wolfe L, Brown MS. Transplacental passage of vancomycin from mother to neonate. Am J Obstet Gynecol 2014; 210:352.e1-352.e4. [PMID: 24679944 DOI: 10.1016/j.ajog.2014.01.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 12/27/2013] [Accepted: 01/13/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The objective of the study was to analyze a large number of patients receiving vancomycin chemoprophylaxis and evaluate the maternal and neonatal cord blood levels at the time of delivery. STUDY DESIGN Every mother who entered labor with a positive group B streptococcal culture and a high-risk penicillin allergy with resistance to clindamycin or unknown sensitivity was consented to participate in the study. In the initial phase of the study, patients received the standard intravenous dose of 1 g every 12 hours. Based on the results, this was changed to a dosing of 15 mg/kg every 12 hours in the second phase and then further modified to 20 mg/kg every 8 hours in the third phase. Maternal and cord blood vancomycin levels were obtained at delivery and evaluated. RESULTS A total of 55 patients consented to participate in the study, with 31 in phase I, 12 in phase II, and 12 in phase III. For the standard-dosing phase I group, only 32% of maternal and 9% of cord blood samples were therapeutic at delivery. For phase II, 50% of maternal and 33% of cord blood values were therapeutic; however, in phase III, 83% of mothers and neonates had therapeutic levels at the time of delivery. CONCLUSION With standard dosing, only 9% of neonates have therapeutic vancomycin levels at delivery. By using a regimen of 20 mg/kg intravenous every 8 hours (maximum individual dose 2 g), the newborn therapeutic level increases above 80%. The pharmacological pattern shows that transplacental passage occurs with fetal levels equaling maternal levels, but transplacental transport is somewhat slow in both directions.
Collapse
|