1
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. A transcriptomic comparison of in vitro models of the human placenta. Placenta 2024; 159:52-61. [PMID: 39637677 DOI: 10.1016/j.placenta.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. The aim of this research was to compare the transcriptomes of common in vitro models of the human placenta compared to bulk human placental tissue. METHODS We performed differential gene expression analysis on publicly available transcriptomic data from 7 in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, Villous Explants, and Trophoblast Stem Cells) and compared to bulk placental tissue from 2 cohort studies (CANDLE and GAPPS) or individual trophoblast cell types derived from bulk placental tissue. RESULTS All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs = 10,624), and the individual trophoblast cell types (Average DEGs = 5413), indicating that there are vast differences in gene expression. Hierarchical clustering identified 54 gene clusters with distinct expression profiles across placental models, with 23 clusters enriched for specific KEGG pathways. Placental cell lines were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as female origin, while JEG-3, JAR, and BeWo cells are of male origin. DISCUSSION None of the models were a close approximation of the human bulk placental transcriptome, highlighting the challenges with model selection. To enable appropriate model selection, we adapted our data into a web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)".
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA.
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Evan J Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA, 98036, USA
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA; Center for Child Health, Behavior and Development, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, 98101, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA, 98195, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| |
Collapse
|
2
|
Lapehn S, Nair S, Firsick EJ, MacDonald J, Thoreson C, Litch JA, Bush NR, Kadam L, Girard S, Myatt L, Prasad B, Sathyanarayana S, Paquette AG. Transcriptomic comparison of in vitro models of the human placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598695. [PMID: 38915703 PMCID: PMC11195179 DOI: 10.1101/2024.06.14.598695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Studying the human placenta through in vitro cell culture methods is necessary due to limited access and amenability of human placental tissue to certain experimental methods as well as distinct anatomical and physiological differences between animal and human placentas. Selecting an in vitro culture model of the human placenta is challenging due to representation of different trophoblast cell types with distinct biological roles and limited comparative studies that define key characteristics of these models. Therefore, the aim of this research was to create a comprehensive transcriptomic comparison of common in vitro models of the human placenta compared to bulk placental tissue from the CANDLE and GAPPS cohorts (N=1083). We performed differential gene expression analysis on publicly available RNA sequencing data from 6 common in vitro models of the human placenta (HTR-8/SVneo, BeWo, JEG-3, JAR, Primary Trophoblasts, and Villous Explants) and compared to CANDLE and GAPPS bulk placental tissue or cytotrophoblast, syncytiotrophoblast, and extravillous trophoblast cell types derived from bulk placental tissue. All in vitro placental models had a substantial number of differentially expressed genes (DEGs, FDR<0.01) compared to the CANDLE and GAPPS placentas (Average DEGs=10,873), and the individual trophoblast cell types (Average DEGs=5,346), indicating that there are vast differences in gene expression compared to bulk and cell-type specific human placental tissue. Hierarchical clustering identified 53 gene clusters with distinct expression profiles across placental models, with 22 clusters enriched for specific KEGG pathways, 7 clusters enriched for high-expression placental genes, and 7 clusters enriched for absorption, distribution, metabolism, and excretion genes. In vitro placental models were classified by fetal sex based on expression of Y-chromosome genes that identified HTR-8/SVneo cells as being of female origin, while JEG-3, JAR, and BeWo cells are of male origin. Overall, none of the models were a close approximation of the transcriptome of bulk human placental tissue, highlighting the challenges with model selection. To enable researchers to select appropriate models, we have compiled data on differential gene expression, clustering, and fetal sex into an accessible web application: "Comparative Transcriptomic Placental Model Atlas (CTPMA)" which can be utilized by researchers to make informed decisions about their selection of in vitro placental models.
Collapse
Affiliation(s)
- Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Sidharth Nair
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - Evan J. Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
| | - Ciara Thoreson
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - James A Litch
- Global Alliance to Prevent Prematurity and Stillbirth, Lynwood, WA 98036 United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences; Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143 United States
| | - Leena Kadam
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Sylvie Girard
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN 55905 United States
| | - Leslie Myatt
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR 97239 United States
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA 99202 United States
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
- Center for Child Health, Behavior and Development, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA 98101 United States
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children!s Research Institute, Seattle, WA 98101 United States
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, Seattle, WA 98195 United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195 United States
| |
Collapse
|
3
|
Marie N, Noble F. Oxycodone, an opioid like the others? Front Psychiatry 2023; 14:1229439. [PMID: 38152360 PMCID: PMC10751306 DOI: 10.3389/fpsyt.2023.1229439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/28/2023] [Indexed: 12/29/2023] Open
Abstract
The over-prescription of opioid analgesics is a growing problem in the field of addiction, which has reached epidemic-like proportions in North America. Over the past decade, oxycodone has gained attention as the leading opioid responsible for the North America opioid crisis. Oxycodone is the most incriminated drug in the early years of the epidemic of opioid use disorder in USA (roughly 1999-2016). The number of preclinical articles on oxycodone is rapidly increasing. Several publications have already compared oxycodone with other opioids, focusing mainly on their analgesic properties. The aim of this review is to focus on the genomic and epigenetic regulatory features of oxycodone compared with other opioid agonists. Our aim is to initiate a discussion of perceptible differences in the pharmacological response observed with these various opioids, particularly after repeated administration in preclinical models commonly used to study drug dependence potential.
Collapse
Affiliation(s)
| | - Florence Noble
- Université Paris Cité, CNRS, Inserm, Pharmacologie et Thérapies des Addictions, Paris, France
| |
Collapse
|
4
|
Pande LJ, Arnet RE, Piper BJ. An Examination of the Complex Pharmacological Properties of the Non-Selective Opioid Modulator Buprenorphine. Pharmaceuticals (Basel) 2023; 16:1397. [PMID: 37895868 PMCID: PMC10610465 DOI: 10.3390/ph16101397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
The goal of this review is to provide a recent examination of the pharmacodynamics as well as pharmacokinetics, misuse potential, toxicology, and prenatal consequences of buprenorphine. Buprenorphine is currently a Schedule III opioid in the US used for opioid-use disorder (OUD) and as an analgesic. Buprenorphine has high affinity for the mu-opioid receptor (MOR), delta (DOR), and kappa (KOR) and intermediate affinity for the nociceptin (NOR). Buprenorphine's active metabolite, norbuprenorphine, crosses the blood-brain barrier, is a potent metabolite that attenuates the analgesic effects of buprenorphine due to binding to NOR, and is responsible for the respiratory depressant effects. The area under the concentration curves are very similar for buprenorphine and norbuprenorphine, which indicates that it is important to consider this metabolite. Crowding sourcing has identified a buprenorphine street value (USD 3.95/mg), indicating some non-medical use. There have also been eleven-thousand reports involving buprenorphine and minors (age < 19) at US poison control centers. Prenatal exposure to clinically relevant dosages in rats produces reductions in myelin and increases in depression-like behavior. In conclusion, the pharmacology of this OUD pharmacotherapy including the consequences of prenatal buprenorphine exposure in humans and experimental animals should continue to be carefully evaluated.
Collapse
Affiliation(s)
- Leana J. Pande
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Touro College of Osteopathic Medicine, Middletown, NY 10027, USA
| | - Rhudjerry E. Arnet
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
| | - Brian J. Piper
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA; (L.J.P.); (R.E.A.)
- Center for Pharmacy Innovation and Outcomes, Danville, PA 17821, USA
| |
Collapse
|
5
|
Chen X, Unadkat JD, Mao Q. Maternal and Fetal Exposure to (-)-Δ 9-tetrahydrocannabinol and Its Major Metabolites in Pregnant Mice Is Differentially Impacted by P-glycoprotein and Breast Cancer Resistance Protein. Drug Metab Dispos 2023; 51:269-275. [PMID: 36446608 PMCID: PMC10029818 DOI: 10.1124/dmd.122.001110] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/13/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
(-)-Δ9-tetrahydrocannabinol (THC) is the primary pharmacological active constituent of cannabis. 11-hydroxy-THC (11-OH-THC) and 11-nor-9-carboxy-THC (THC-COOH) are respectively the active and nonactive circulating metabolites of THC in humans. While previous animal studies reported that THC could be a substrate of mouse P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), we have shown, in vitro, that only THC-COOH is a weak substrate of human BCRP, but not of P-gp. To confirm these findings and to investigate the role of P-gp and/or Bcrp in the maternal-fetal disposition of THC and its metabolites, we administrated 3 mg/kg of THC retro-orbitally to FVB wild-type (WT), P-gp -/-, Bcrp -/-, or P-gp-/- /Bcrp-/- pregnant mice on gestation day 18 and estimated the area under the concentration-time curve (AUC) of the cannabinoids in the maternal plasma, maternal brain, placenta, and fetus, as well as the tissue/maternal plasma AUC geometric mean ratios (GMRs) using a pooled data bootstrap approach. We found that the dose-normalized maternal plasma AUCs of THC in P-gp-/- and P-gp-/- /Bcrp-/- mice, and the placenta-to-maternal plasma AUC GMR of THC in Bcrp-/- mice were 279%, 271%, and 167% of those in WT mice, respectively. Surprisingly, the tissue-to-maternal plasma AUC GMRs of THC and its major metabolites in the maternal brain, placenta, or fetus in P-gp -/-, Bcrp -/- or P-gp-/- /Bcrp-/- mice were 28-78% of those in WT mice. This study revealed that P-gp and Bcrp do not play a role in limiting maternal brain and fetal exposure to THC and its major metabolites in pregnant mice. SIGNIFICANCE STATEMENT: This study systematically investigated whether P-gp and/or Bcrp in pregnant mice can alter the disposition of THC, 11-OH-THC, and THC-COOH. Surprisingly, except for Bcrp, which limits placental (but not fetal) exposure to THC, we found that P-gp-/- , Bcrp-/- , and/or P-gp-/- /Bcrp-/- significantly decreased exposure to THC and/or its metabolites in maternal brain, placenta, or fetus. The mechanistic basis for this decrease is unclear and needs further investigation. If replicated in humans, P-gp- or BCRP-based drug-cannabinoid interactions are not of concern.
Collapse
MESH Headings
- Pregnancy
- Mice
- Female
- Humans
- Animals
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Dronabinol/metabolism
- Placenta/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP-Binding Cassette Transporters/metabolism
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Breast Neoplasms/metabolism
Collapse
Affiliation(s)
- Xin Chen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| |
Collapse
|
6
|
Torres-Vergara P, Rivera R, Escudero C, Penny J. Maternal and Fetal Expression of ATP-Binding Cassette and Solute Carrier Transporters Involved in the Brain Disposition of Drugs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:149-177. [PMID: 37466773 DOI: 10.1007/978-3-031-32554-0_7] [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: 07/20/2023]
Abstract
Evidence from preclinical and clinical studies demonstrate that pregnancy is a physiological state capable of modifying drug disposition. Factors including increased hepatic metabolism and renal excretion are responsible for impacting disposition, and the role of membrane transporters expressed in biological barriers, including the placental- and blood-brain barriers, has received considerable attention. In this regard, the brain disposition of drugs in the mother and fetus has been the subject of studies attempting to characterize the mechanisms by which pregnancy could alter the expression of ATP-binding cassette (ABC) and solute carrier (SLC) transporters. This chapter will summarize findings of the influence of pregnancy on the maternal and fetal expression of ABC and SLC transporters in the brain and the consequences of such changes on the disposition of therapeutic drugs.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile.
| | - Robin Rivera
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile
- Laboratorio de Fisiología Vascular, Facultad de Ciencias Básicas, Universidad del Bio Bio, Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| |
Collapse
|
7
|
Mao Q, Chen X. An update on placental drug transport and its relevance to fetal drug exposure. MEDICAL REVIEW (2021) 2022; 2:501-511. [PMID: 37724167 PMCID: PMC10388746 DOI: 10.1515/mr-2022-0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/27/2022] [Indexed: 09/20/2023]
Abstract
Pregnant women are often complicated with diseases that require treatment with medication. Most drugs administered to pregnant women are off-label without the necessary dose, efficacy, and safety information. Knowledge concerning drug transfer across the placental barrier is essential for understanding fetal drug exposure and hence drug safety and efficacy to the fetus. Transporters expressed in the placenta, including adenosine triphosphate (ATP)-binding cassette efflux transporters and solute carrier uptake transporters, play important roles in determining drug transfer across the placental barrier, leading to fetal exposure to the drugs. In this review, we provide an update on placental drug transport, including in vitro cell/tissue, ex vivo human placenta perfusion, and in vivo animal studies that can be used to determine the expression and function of drug transporters in the placenta as well as placental drug transfer and fetal drug exposure. We also describe how the knowledge of placental drug transfer through passive diffusion or active transport can be combined with physiologically based pharmacokinetic modeling and simulation to predict systemic fetal drug exposure. Finally, we highlight knowledge gaps in studying placental drug transport and predicting fetal drug exposure and discuss future research directions to fill these gaps.
Collapse
Affiliation(s)
- Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Xin Chen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| |
Collapse
|
8
|
Balhara A, Kumar AR, Unadkat JD. Predicting Human Fetal Drug Exposure Through Maternal-Fetal PBPK Modeling and In Vitro or Ex Vivo Studies. J Clin Pharmacol 2022; 62 Suppl 1:S94-S114. [PMID: 36106781 PMCID: PMC9494623 DOI: 10.1002/jcph.2117] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/20/2022] [Indexed: 11/06/2022]
Abstract
Medication (drug) use in human pregnancy is prevalent. Determining fetal safety and efficacy of drugs is logistically challenging. However, predicting (not measuring) fetal drug exposure (systemic and tissue) throughout pregnancy is possible through maternal-fetal physiologically based pharmacokinetic (PBPK) modeling and simulation. Such prediction can inform fetal drug safety and efficacy. Fetal drug exposure can be quantified in 2 complementary ways. First, the ratio of the steady-state unbound plasma concentration in the fetal plasma (or area under the plasma concentration-time curve) to the corresponding maternal plasma concentration (ie, Kp,uu ). Second, the maximum unbound peak (Cu,max,ss,f ) and trough (Cu,min,ss,f ) fetal steady-state plasma concentrations. We (and others) have developed a maternal-fetal PBPK model that can successfully predict maternal drug exposure. To predict fetal drug exposure, the model needs to be populated with drug specific parameters, of which transplacental clearances (active and/or passive) and placental/fetal metabolism of the drug are critical. Herein, we describe in vitro studies in cells/tissue fractions or the perfused human placenta that can be used to determine these drug-specific parameters. In addition, we provide examples whereby this approach has successfully predicted systemic fetal exposure to drugs that passively or actively cross the placenta. Apart from maternal-fetal PBPK models, animal studies also have the potential to estimate fetal drug exposure by allometric scaling. Whether such scaling will be successful is yet to be determined. Here, we review the above approaches to predict fetal drug exposure, outline gaps in our knowledge to make such predictions and map out future research directions that could fill these gaps.
Collapse
Affiliation(s)
- Ankit Balhara
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Aditya R Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Hakomäki H, Eskola S, Kokki H, Lehtonen M, Räsänen J, Laaksonen S, Voipio HM, Ranta VP, Kokki M. Central Nervous System Distribution of Buprenorphine in Pregnant Sheep, Fetuses and Newborn Lambs After Continuous Transdermal and Single Subcutaneous Extended-Release Dosing. Eur J Pharm Sci 2022; 178:106283. [PMID: 36029997 DOI: 10.1016/j.ejps.2022.106283] [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: 05/24/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022]
Abstract
Buprenorphine is used during pregnancy for the treatment of opioid use disorder. Limited data exist on the central nervous system (CNS) permeation and distribution, and on the fetal exposure to buprenorphine. The aim of our study was to determine the extent of buprenorphine distribution to CNS in the pregnant sheep, and their fetus at steady-state, and their newborn lambs postdelivery, using three different dosing regimens. Twenty-eight pregnant ewes in late gestation received buprenorphine via 7-day transdermal patch releasing buprenorphine 20 µg/h (n=9) or 40 µg/h (n=11), or an extended-release 8 mg/week subcutaneous injection (n=8). Plasma, cerebrospinal fluid, and CNS tissue samples were collected at steady-state from ewes and fetuses, and from lambs 0.33 - 45 hours after delivery. High accumulation of buprenorphine was observed in all CNS tissues. The median CNS/plasma concentration -ratios of buprenorphine in different CNS areas ranged between 13 and 50 in the ewes, and between 26 and 198 in the fetuses. In the ewes the CNS/plasma -ratios were similar after the three dosing regimens, but higher in the fetuses in the 40 µg/h dosing group, medians 65 - 122, than in the 20 µg/h group, medians 26 - 54. The subcutaneous injection (theoretical release rate 47.6 µg/h) produced higher concentrations than observed after 40 µg/h transdermal patch dosing. The median fetal/maternal concentration -ratios in different dosing groups ranged between 0.21 and 0.54 in plasma, and between 0.38 and 1.3 in CNS tissues, respectively, with the highest ratios observed in the spinal cord. Buprenorphine concentrations in the cerebrospinal fluid were 8 - 13 % of the concurrent plasma concentration in the ewes and 28 % in the fetuses. Buprenorphine was quantifiable in the newborn lambs' plasma and CNS tissues two days postdelivery. Norbuprenorphine was analyzed from all plasma, cerebrospinal fluid, and CNS tissue samples but was nondetectable or below the LLOQ in most. The current study demonstrates that buprenorphine accumulates into CNS tissues at much higher concentrations than in plasma in pregnant sheep, fetuses, and their newborn lambs even 45 hours after delivery.
Collapse
Key Words
- BUP, Buprenorphine
- CL, Plasma clearance
- CNS, Central nervous system
- CSF, Cerebrospinal fluid
- F/M -ratio, Fetal to maternal concentration ratio
- HPLC, , High-performance liquid chromatography
- L/M -ratio, Lamb to maternal concentration ratio
- LC/MS/MS, Liquid chromatography - tandem mass spectrometry
- LLOQ, Lower limit of quantification
- NBUP, Norbuprenorphine
- brain
- buprenorphine
- pharmacokinetics, pregnancy
- sheep
- tissue
Collapse
Affiliation(s)
| | - Sophia Eskola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Hannu Kokki
- School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Juha Räsänen
- Fetal Medicine Center, Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sakari Laaksonen
- Department of Comparative Medicine, Oulu Laboratory Animal Centre, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Hanna-Marja Voipio
- Department of Comparative Medicine, Oulu Laboratory Animal Centre, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Veli-Pekka Ranta
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Merja Kokki
- Department of Anesthesiology and Intensive Care, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
10
|
Eapen-John D, Mohiuddin AG, Kennedy JL. A potential paradigm shift in opioid crisis management: The role of pharmacogenomics. World J Biol Psychiatry 2022; 23:411-423. [PMID: 34854362 DOI: 10.1080/15622975.2021.2012397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pharmacogenetic investigations into the opioid crisis suggest genetic variation could be a significant cause of opioid-related morbidity and mortality. Variability in opioid system genes, including single nucleotide polymorphisms, manifest after pharmacogenetic testing, as previously invisible risk factors for addiction and overdose. Pharmacodynamic genes regulate opioid-sensitive brain networks and neural reward circuitry. Pharmacokinetic genes expressed in drug metabolic pathways regulate blood levels of active vs. inactive opioid metabolites. Elucidating the complex interplay of genetic variations in pharmacokinetic and pharmacodynamic pathways will shed new light on the addictive and toxic properties of opioids. This narrative review serves to promote understanding of key genetic mechanisms affecting the metabolism and actions of opioids, and to explore causes of the recent surge in opioid-related mortality associated with COVID-19. Personalised treatment plans centred around an individual's genetic makeup could make opioid-based pain management and opioid use disorder (OUD) treatments safer and more effective at both the individual and system levels.
Collapse
Affiliation(s)
- David Eapen-John
- Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Ayeshah G Mohiuddin
- Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James L Kennedy
- Molecular Brain Science, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada
| |
Collapse
|
11
|
Fujita A, Noguchi S, Hamada R, Inoue S, Shimada T, Katakura S, Maruyama T, Sai Y, Nishimura T, Tomi M. Limited Impact of Murine Placental MDR1 on Fetal Exposure of Certain Drugs Explained by Bypass Transfer Between Adjacent Syncytiotrophoblast Layers. Pharm Res 2022; 39:1645-1658. [PMID: 35083640 PMCID: PMC9246986 DOI: 10.1007/s11095-022-03165-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022]
Abstract
Purpose Multidrug resistance protein 1 (MDR1) is located at the interface between two syncytiotrophoblast layers in rodent placenta, and may influence fetal drug distribution. Here, we quantitatively compare the functional impact per single MDR1 molecule of MDR1 at the placental barrier and blood-brain barrier in mice. Methods MDR1A and MDR1B proteins were quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Paclitaxel or digoxin was continuously administered to pregnant Mdr1a−/−/Mdr1b−/− or wild-type mice, and the drug concentrations in the maternal and fetal plasma and maternal brain were quantified by LC-MS/MS. Results MDR1A and MDR1B proteins are expressed in the membrane of mouse placental labyrinth, and total MDR1 at the placental barrier amounts to about 30% of that at the blood-brain barrier. The fetal-to-maternal plasma concentration ratio of digoxin was only marginally affected in Mdr1a−/−/Mdr1b−/− mice, while that of paclitaxel showed a several-fold increase. No such difference between the two drugs was found in the maternal brain distribution. The impact per single MDR1 molecule on the fetal distribution of digoxin was calculated to be much lower than that on the brain distribution, but this was not the case for paclitaxel. Our pharmacokinetic model indicates that the impact of placental MDR1 is inversely correlated to the ratio of permeability through gap junctions connecting the two syncytiotrophoblast layers to passive diffusion permeability. Conclusion Our findings indicate that murine placental MDR1 has a minimal influence on the fetal concentration of certain substrates, such as digoxin, due to bypass transfer, probably via connexin26 gap junctions. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-022-03165-6.
Collapse
Affiliation(s)
- Arimi Fujita
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.,Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan.,Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Saki Noguchi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Rika Hamada
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Satoko Inoue
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Tsutomu Shimada
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan.,Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Satomi Katakura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tetsuo Maruyama
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshimichi Sai
- Department of Clinical Pharmacokinetics, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan.,Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Tomohiro Nishimura
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Masatoshi Tomi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
12
|
López Quiñones AJ, Shireman LM, Wang J. Development and validation of a LC-MS/MS method for in vivo quantification of meta-iodobenzylguanidine (mIBG). J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1181:122927. [PMID: 34530306 DOI: 10.1016/j.jchromb.2021.122927] [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: 04/29/2021] [Revised: 08/03/2021] [Accepted: 09/01/2021] [Indexed: 10/20/2022]
Abstract
meta-iodobenzylguanidine (mIBG) is a radiopharmaceutical used for the diagnosis and treatment of neuroendocrine cancers. Previous quantification of mIBG in biodistribution and pharmacokinetic studies mainly relied on the use of radiolabeled mIBG, which involves the handling of highly radioactive materials. The goal of this study was to develop a nonradioactive analytical method for quantifying mIBG in mouse plasma and tissue homogenates using high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS). Samples were prepared for analysis using a protein precipitation method. Mass spectrometry analysis was performed using 4-hydroxyphenformin as the internal standard, and the mass-to-charge transitions were 276.1 → 217.0 for mIBG and 222.1 → 121.0 for 4-hydroxyphenformin. The quantification limit of mIBG was 0.98 ng/mL, and the method was linear up to 500 ng/mL. The accuracy, inter-day and intra-day precision were 96-112%, 5.5-14.4%, and 3.7-14.1%, respectively, suggesting that the method was accurate and precise in quantifying mIBG at multiple concentrations in mouse plasma and liver homogenates. The extraction recovery was 96-106% and the matrix effect was 95-110%, indicating that the method was reproducible in quantifying mIBG with minimal impact from the biological matrices. In summary, we have developed and validated a fast, high-throughput quantification method of non-radiolabeled mIBG using LC-MS/MS. This method is reproducible, accurate, and precise, and can be used to quantify mIBG in plasma and tissue matrices to determine the pharmacokinetics and biodistribution of mIBG in preclinical animal models.
Collapse
Affiliation(s)
| | - Laura M Shireman
- Department of Pharmaceutics, University of Washington, Seattle WA 98195, United States
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Seattle WA 98195, United States.
| |
Collapse
|
13
|
Bishop-Freeman SC, Friederich LW, Feaster MS, Hudson JS. Buprenorphine-Related Deaths in North Carolina from 2010-2018. J Anal Toxicol 2021; 45:780-791. [PMID: 34145443 DOI: 10.1093/jat/bkab073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
Buprenorphine is a commonly prescribed medication for the treatment of opioid-use disorder. As prescriptions increase in North Carolina, buprenorphine is more frequently encountered statewide in routine postmortem casework. Between 2010 and 2018, there were 131 select cases investigated by the Office of the Chief Medical Examiner where buprenorphine was detected in peripheral blood and considered a primary cause of death, with no other opioids present and no other non-opioid substances found in the lethal range. The decedents ranged in age from 14 to 64 years, with 67% male. The mean/median peripheral blood concentrations were 4.1/2.1 ng/mL for buprenorphine and 7.8/3.4 ng/mL for the metabolite, norbuprenorphine. These postmortem blood concentrations overlap antemortem therapeutic concentrations in plasma reported in the literature for opioid-dependent subjects receiving sublingual maintenance therapy. The pathologist considered scene findings, prescription history, autopsy findings, toxicological analysis, and decedent behavior prior to death to conclude a drug-related cause of death. Many of the deaths were complicated by the presence of other central nervous system depressants along with contributory underlying cardiovascular and respiratory disease. The three most prevalent additive substances were alprazolam, ethanol, and gabapentin, found in 67, 36, and 32 cases out of 131, respectively. Interpreting buprenorphine involvement in a death is complex, and instances may be under-estimated in epidemiological data because of the lack of a defined toxic or lethal range in postmortem blood along with its good safety profile. As expansion to access of opioid-use disorder treatment becomes a priority, awareness of the challenges of postmortem interpretation is needed as increased use and diversion of buprenorphine are inevitable.
Collapse
Affiliation(s)
- Sandra C Bishop-Freeman
- Office of the Chief Medical Examiner, Raleigh, NC, USA.,UNC Department of Pathology and Laboratory Medicine, Chapel Hill, NC, USA
| | | | | | | |
Collapse
|
14
|
Shum S, Isoherranen N. Human Fetal Liver Metabolism of Oxycodone Is Mediated by CYP3A7. AAPS J 2021; 23:24. [PMID: 33438174 PMCID: PMC8106324 DOI: 10.1208/s12248-020-00537-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/19/2020] [Indexed: 11/30/2022] Open
Abstract
Oxycodone is an opioid analgesic that is commonly prescribed to pregnant women to treat moderate-to-severe pain. It has been shown to cross the placenta and distribute to the fetus. Oxycodone is mainly metabolized by CYP3A4 in the adult liver. Since CYP3A7 is abundantly expressed in the fetal liver and has overlapping substrate specificity with CYP3A4, we hypothesized that the fetal liver may significantly limit fetal exposure to oxycodone. This study showed that oxycodone is metabolized by CYP3A7 to noroxycodone in fetal liver microsomes (FLMs). The measured CYP3A7 expression was 191-409 pmol/mg protein in 14 FLMs, and an intersystem extrapolation factor (ISEF) for CYP3A7 was 0.016-0.066 in the panel of fetal livers using 6β-OH-testosterone formation as the probe reaction. Noroxycodone formation in the fetal liver was predicted from formation rate by recombinant CYP3A7, CYP3A7 expression level and the established ISEF value with average fold error of 1.25. Based on the intrinsic clearance of oxycodone measured in FLM, the fetal hepatic clearance (CLh) at term was predicted to be 495 (range: 66.4-936) μL/min, a value that is > 99% lower than the predicted adult liver CLh. The predicted fetal hepatic extraction ratio was 0.0019 (range: 0.00003-0.0036). These results suggest that fetal liver metabolism does not quantitatively contribute to the total systemic clearance of oxycodone in pregnant women nor does it provide a barrier for limiting fetal exposure to oxycodone. Additionally, since CYP3A7 forms noroxycodone, an inactive metabolite, the metabolism in the fetal liver is unlikely to affect fetal opioid activity.
Collapse
Affiliation(s)
- Sara Shum
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
- University of Washington, Health Science Building Room H-272M, Box 357610, Seattle, Washington, USA.
| |
Collapse
|
15
|
A review of the existing literature on buprenorphine pharmacogenomics. THE PHARMACOGENOMICS JOURNAL 2020; 21:128-139. [PMID: 33154520 DOI: 10.1038/s41397-020-00198-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022]
Abstract
Buprenorphine is an effective treatment for opioid dependence; however, it demonstrates individual variability in efficacy. Pharmacogenomics may explain this drug response variability and could allow for tailored therapy on an individual basis. The Food and Drug Administration and the Clinical Pharmacogenomics Implementation Consortium have guidelines on pharmacogenomic testing for some opioids (e.g., codeine); however, no guidelines exist for the partial opioid agonist buprenorphine. Pharmacogenomic testing targets for buprenorphine include pharmacodynamic genes like the mu-opioid receptor (MOP receptor) and catechol-O-methyltransferase (COMT), as well as the pharmacokinetic genes like the CYP enzymes. In this review we identified genotypes in patients with opioid addiction receiving buprenorphine that may result in altered therapeutic dosing and increased rate of relapse. The OPRM1 A118G single nucleotide polymorphism (SNP rs1799971) gene variant encoding the N40D MOP receptor has been associated with variable efficacy and response to treatment in both adult and neonatal patients receiving buprenorphine for treatment of opioid withdrawal. An SNP associated with rs678849 of OPRD1, coding for the delta opioid receptor, was associated with opioid relapse as indicated by opioid positive urine drug screens; there was also sex specific SNP identified at rs581111 and rs529520 in the European American population. COMT variability, particularly in rs4680, has been associated with length of stay and need for opioid treatment in patients with neonatal abstinence syndrome. Variations of the pharmacokinetic gene for CYP3A4 showed that the ultrarapid metabolizer phenotype required higher doses of buprenorphine. Genotyping of patients may allow us to appropriately tailor buprenorphine therapy to individual patients and lead to improved patient outcomes; however, further research on the pharmacogenomics of buprenorphine is needed.
Collapse
|
16
|
Bicker J, Alves G, Falcão A, Fortuna A. Timing in drug absorption and disposition: The past, present, and future of chronopharmacokinetics. Br J Pharmacol 2020; 177:2215-2239. [PMID: 32056195 DOI: 10.1111/bph.15017] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/05/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The importance of drug dosing time in pharmacokinetics, pharmacodynamics, and toxicity is receiving increasing attention from the scientific community. In spite of mounting evidence that circadian oscillations affect drug absorption, distribution, metabolism, and excretion (ADME), there remain many unanswered questions in this field and, occasionally, conflicting experimental results. Such data arise not only from translational difficulties caused by interspecies differences but also from variability in study design and a lack of understanding of how the circadian clock affects physiological factors that strongly influence ADME, namely, the expression and activity of drug transporters. Hence, the main goal of this review is to provide an updated analysis of the role of the circadian rhythm in drug absorption, distribution across blood-tissue barriers, metabolism in hepatic and extra-hepatic tissues, and hepatobiliary and renal excretion. It is expected that the research suggestions proposed here will contribute to a tissue-targeted and time-targeted pharmacotherapy.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Gilberto Alves
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CIBIT/ICNAS-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
17
|
Bergman L, Torres-Vergara P, Penny J, Wikström J, Nelander M, Leon J, Tolcher M, Roberts JM, Wikström AK, Escudero C. Investigating Maternal Brain Alterations in Preeclampsia: the Need for a Multidisciplinary Effort. Curr Hypertens Rep 2019; 21:72. [PMID: 31375930 DOI: 10.1007/s11906-019-0977-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW To provide insight into the mechanisms underlying cerebral pathophysiology and to highlight possible methods for evaluation, screening, and surveillance of cerebral complications in preeclampsia. RECENT FINDINGS The pathophysiology of eclampsia remains enigmatic. Animal studies show that the cerebral circulation in pregnancy and preeclampsia might be affected with increased permeability over the blood-brain barrier and altered cerebral blood flow due to impaired cerebral autoregulation. The increased blood pressure cannot be the only underlying cause of eclampsia and cerebral edema, since some cases of eclampsia arise without simultaneous hypertension. Findings from animal studies need to be confirmed in human tissues. Evaluation of brain alterations in preeclampsia and eclampsia is challenging and demands a multidisciplinary collaboration, since no single method can accurately and fully describe how preeclampsia affects the brain. Cerebral complications of preeclampsia are significant factors in maternal morbidity and mortality worldwide. No single method can accurately describe the full picture of how preeclampsia affects the brain vasculature and parenchyma. We recommend an international and multidisciplinary effort not only to overcome the issue of limited sample availability but also to optimize the quality of research.
Collapse
Affiliation(s)
- Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.
- Center for Clinical Research Dalarna, Falun, Uppsala, Sweden.
| | - Pablo Torres-Vergara
- Pharmacy Department, Faculty of Pharmacy, Universidad de Concepción, Concepción, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Johan Wikström
- Department of Radiology, Uppsala University, Uppsala, Sweden
| | - Maria Nelander
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Jose Leon
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis, (LFV-GIANT), Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile
| | - Mary Tolcher
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - James M Roberts
- Magee Womens Research Institute, Dept of Obstetrics Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anna-Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis, (LFV-GIANT), Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán, Chile.
| |
Collapse
|
18
|
Nicolas JM, de Lange ECM. Mind the Gaps: Ontogeny of Human Brain P-gp and Its Impact on Drug Toxicity. AAPS JOURNAL 2019; 21:67. [PMID: 31140038 DOI: 10.1208/s12248-019-0340-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/10/2019] [Indexed: 12/18/2022]
Abstract
Available data on human brain P-glycoprotein ontogeny during infancy and childhood are limited. This review discusses the current body of data relating to maturation of human brain P-glycoprotein including transporter expression levels in post-mortem human brain samples, in vivo transporter activity using probe substrates, surrogate marker endpoints, and extrapolations from animal models. Overall, the data tend to confirm that human brain P-glycoprotein activity keeps developing after birth, although with a developmental time frame that remains unclear. This knowledge gap is a concern given the critical role of brain P-glycoprotein in drug safety and efficacy, and the vulnerable nature of the pediatric population. Future research could include the measurement of brain P-glycoprotein activity across age groups using positron emission tomography or central pharmacodynamic responses. For now, caution is advised when extrapolating adult data to children aged younger than 2 years for drugs with P-glycoprotein-dependent central nervous system activity.
Collapse
Affiliation(s)
- Jean-Marie Nicolas
- Quantitative Pharmacology DMPK Department, UCB BioPharma, Chemin du Foriest, 1420, Braine L'Alleud, Belgium.
| | - Elizabeth C M de Lange
- Research Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| |
Collapse
|
19
|
Torres-Vergara P, Escudero C, Penny J. Drug Transport at the Brain and Endothelial Dysfunction in Preeclampsia: Implications and Perspectives. Front Physiol 2018; 9:1502. [PMID: 30459636 PMCID: PMC6232255 DOI: 10.3389/fphys.2018.01502] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/05/2018] [Indexed: 12/20/2022] Open
Abstract
Transport of drugs across biological barriers has been a subject of study for decades. The discovery and characterization of proteins that confer the barrier properties of endothelia and epithelia, including tight junction proteins and membrane transporters belonging to the ATP-binding cassette (ABC) and Solute Carrier (SLC) families, represented a significant step forward into understanding the mechanisms that govern drug disposition. Subsequently, numerous studies, including both pre-clinical approaches and clinical investigations, have been carried out to determine the influence of physiological and pathological states on drug disposition. Importantly, there has been increasing interest in gaining a better understanding of drug disposition during pregnancy, since epidemiological and clinical studies have demonstrated that the use of medications by pregnant women is significant and this condition embodies a series of significant anatomical and physiological modifications, particularly at excretory organs and barrier sites (e.g., placenta, breast) expressing transporter proteins which influence pharmacokinetics. Currently, most of the research in this field has focused on the expression profiling of transporter proteins in trophoblasts and endothelial cells of the placenta, regulation of drug-resistance mechanisms in disease states and pharmacokinetic studies. However, little attention has been placed on the influence that the cerebrovascular dysfunction present in pregnancy-related disorders, such as preeclampsia, might exert on drug disposition in the mother’s brain. This issue is particularly important since recent findings have demonstrated that preeclamptic women suffer from long-term alterations in the integrity of the blood-brain barrier (BBB). In this review we aim to analyze the available evidence regarding the influence of pregnancy on the expression of transporters and TJ proteins in brain endothelial cells, as well the mechanisms that govern the pathophysiological alterations in the BBB of women who experience preeclampsia. Future research efforts should be focused not only on achieving a better understanding of the influence of preeclampsia-associated endothelial dysfunction on drug disposition, but also in optimizing the pharmacological treatments of women suffering pregnancy-related disorders, its comorbidities and to develop new therapies aiming to restore the integrity of the BBB.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Concepción, Chile.,Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.,Vascular Physiology Laboratory, Department of Basic Sciences, Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile.,Red Iberoamericana de Alteraciones Vasculares Asociadas a Trastornos del Embarazo (RIVA-TREM), Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
20
|
Han LW, Gao C, Mao Q. An update on expression and function of P-gp/ABCB1 and BCRP/ABCG2 in the placenta and fetus. Expert Opin Drug Metab Toxicol 2018; 14:817-829. [PMID: 30010462 DOI: 10.1080/17425255.2018.1499726] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION P-glycoprotein (P-gp)/ABCB1 and breast cancer resistance protein (BCRP)/ABCG2 are highly expressed in the placenta and fetus throughout gestation and can modulate exposure and toxicity of drugs and xenobiotics to the vulnerable fetus during the sensitive times of growth and development. We aim to provide an update on current knowledge on placental and fetal expressions of the two transporters in different species, and to provide insight on interpreting transporter expression and fetal exposure relative to the concept of fraction of drug transported. Areas covered: Comprehensive literature review through PubMed (primarily from July 2010 to February 2018) on P-gp and BCRP expression and function in the placenta and fetus of primarily human, mouse, rat, and guinea pig. Expert opinion: While there are many commonalities in the expression and function of P-gp and BCRP in the placenta and fetal tissues across species, there are distinct differences in expression levels and temporal changes. Further studies are needed to quantify protein abundance of these transporters and functionally assess their activities at various gestational stages. Combining the knowledge of interspecies differences and the concept of fraction of drug transported, we may better predict the magnitude of impact these transporters have on fetal drug exposure.
Collapse
Affiliation(s)
- Lyrialle W Han
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| | - Chunying Gao
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| | - Qingcheng Mao
- a Department of Pharmaceutics, School of Pharmacy , University of Washington , Seattle , WA , USA
| |
Collapse
|
21
|
Liao MZ, Gao C, Bhatt DK, Prasad B, Mao Q. Quantitative Proteomics Reveals Changes in Transporter Protein Abundance in Liver, Kidney and Brain of Mice by Pregnancy. Drug Metab Lett 2018; 12:145-152. [PMID: 29938623 PMCID: PMC6350206 DOI: 10.2174/1872312812666180625122810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/11/2022]
Abstract
Background: Few studies have systematically investigated pregnancy-induced changes in protein abundance of drug transporters in organs important for drug/xenobiotic disposition. Objective: The goal of this study was to compare protein abundance of important drug/xenobiotic trans-porters including Abcb1a, Abcg2, Abcc2, and Slco1b2 in the liver, kidney and brain of pregnant mice on gestation day 15 to that of non-pregnant mice. Methods: The mass spectrometry-based proteomics was used to quantify changes in protein abundance of transporters in tissues from pregnant and non-pregnant mice. Results: The protein levels of hepatic Abcc2, Abcc3, and Slco1a4 per µg of total membrane proteins were significantly decreased by pregnancy by 24%, 72%, and 70%, respectively. The protein levels of Abcg2, Abcc2, and Slco2b1 per µg of total membrane proteins in the kidney were significantly decreased by pregnancy by 43%, 50%, and 46%, respectively. After scaling to the whole liver with consideration of increase in liver weight in pregnant mice, the protein abundance of Abcb1a, Abcg2, Abcc2, Abcb11, Abcc4, Slco1a1, and Slco1b2 in the liver was ~50-100% higher in pregnant mice, while those of Abcc3 and Slco1a4 were ~40% lower. After scaling to the whole kidney, none of the transporters examined were significantly changed by pregnancy. Only Abcg2 and Abcb1a were quantifiable in the brain and their abundance in the brain was not influenced by pregnancy. Conclusion: Protein abundance of drug transporters can be significantly changed particularly in the liver by pregnancy. These results will be helpful to understand pregnancy-induced changes in drug/xenobiotic disposition in the mouse model.
Collapse
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington DC, 98195, United States
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington DC, 98195, United States
| | - Deepak Kumar Bhatt
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington DC, 98195, United States
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington DC, 98195, United States
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington DC, 98195, United States
| |
Collapse
|
22
|
Liao MZ, Gao C, Phillips BR, Neradugomma NK, Han LW, Bhatt DK, Prasad B, Shen DD, Mao Q. Pregnancy Increases Norbuprenorphine Clearance in Mice by Induction of Hepatic Glucuronidation. Drug Metab Dispos 2017; 46:100-108. [PMID: 29158248 DOI: 10.1124/dmd.117.076745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/17/2017] [Indexed: 01/09/2023] Open
Abstract
Norbuprenorphine (NBUP) is the major active metabolite of buprenorphine (BUP) that is commonly used to treat opiate addiction during pregnancy; it possesses 25% of BUP's analgesic activity and 10 times BUP's respiratory depression effect. To optimize BUP's dosing regimen during pregnancy with better efficacy and safety, it is important to understand how pregnancy affects NBUP disposition. In this study, we examined the pharmacokinetics of NBUP in pregnant and nonpregnant mice by administering the same amount of NBUP through retro-orbital injection. We demonstrated that the systemic clearance (CL) of NBUP in pregnant mice increased ∼2.5-fold compared with nonpregnant mice. Intrinsic CL of NBUP by glucuronidation in mouse liver microsomes from pregnant mice was ∼2 times greater than that from nonpregnant mice. Targeted liquid chromatography tandem-mass spectrometry proteomics quantification revealed that hepatic Ugt1a1 and Ugt2b1 protein levels in the same amount of total liver membrane proteins were significantly increased by ∼50% in pregnant mice versus nonpregnant mice. After scaling to the whole liver with consideration of the increase in liver protein content and liver weight, we found that the amounts of Ugt1a1, Ugt1a10, Ugt2b1, and Ugt2b35 protein in the whole liver of pregnant mice were significantly increased ∼2-fold compared with nonpregnant mice. These data suggest that the increased systemic CL of NBUP in pregnant mice is likely caused by an induction of hepatic Ugt expression and activity. The data provide a basis for further mechanistic analysis of pregnancy-induced changes in the disposition of NBUP and drugs that are predominately and extensively metabolized by Ugts.
Collapse
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Chunying Gao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Brian R Phillips
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Naveen K Neradugomma
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Lyrialle W Han
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Deepak Kumar Bhatt
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Bhagwat Prasad
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
23
|
Bhatt DK, Prasad B. Critical Issues and Optimized Practices in Quantification of Protein Abundance Level to Determine Interindividual Variability in DMET Proteins by LC-MS/MS Proteomics. Clin Pharmacol Ther 2017; 103:619-630. [PMID: 28833066 DOI: 10.1002/cpt.819] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 08/12/2017] [Indexed: 12/16/2022]
Abstract
Protein quantification data on drug metabolizing enzymes and transporters (collectively referred as DMET proteins) in human tissues are useful in predicting interindividual variability in drug disposition. While targeted proteomics is an emerging technique for quantification of DMET proteins, the methodology involves significant technical challenges especially when multiple samples are analyzed in a single study over a long period of time. Therefore, it is important to thoroughly address the critical variables that could affect DMET protein quantification.
Collapse
Affiliation(s)
- Deepak Kumar Bhatt
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
24
|
Liao MZ, Shen DD, Mao Q. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine-Response to 'Letter to the Editor'. Pharmacol Res 2017; 121:253. [PMID: 28411002 DOI: 10.1016/j.phrs.2017.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/06/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Michael Z Liao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Danny D Shen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Qingcheng Mao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
25
|
Dash RP, Rais R. P-gp/ABCB1 exerts differential impacts on brain and fetal exposure to norbuprenorphine-Significance of zeitgeber time on pharmacokinetics. Pharmacol Res 2017; 121:251-252. [PMID: 28400151 DOI: 10.1016/j.phrs.2017.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Ranjeet Prasad Dash
- Drug Metabolism and Pharmacokinetics, Johns Hopkins Drug Discovery Program, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rana Rais
- Drug Metabolism and Pharmacokinetics, Johns Hopkins Drug Discovery Program, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|