1
|
Lu Z, Guo Y, Xu D, Xiao H, Dai Y, Liu K, Chen L, Wang H. Developmental toxicity and programming alterations of multiple organs in offspring induced by medication during pregnancy. Acta Pharm Sin B 2023; 13:460-477. [PMID: 36873163 PMCID: PMC9978644 DOI: 10.1016/j.apsb.2022.05.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Medication during pregnancy is widespread, but there are few reports on its fetal safety. Recent studies suggest that medication during pregnancy can affect fetal morphological and functional development through multiple pathways, multiple organs, and multiple targets. Its mechanisms involve direct ways such as oxidative stress, epigenetic modification, and metabolic activation, and it may also be indirectly caused by placental dysfunction. Further studies have found that medication during pregnancy may also indirectly lead to multi-organ developmental programming, functional homeostasis changes, and susceptibility to related diseases in offspring by inducing fetal intrauterine exposure to too high or too low levels of maternal-derived glucocorticoids. The organ developmental toxicity and programming alterations caused by medication during pregnancy may also have gender differences and multi-generational genetic effects mediated by abnormal epigenetic modification. Combined with the latest research results of our laboratory, this paper reviews the latest research progress on the developmental toxicity and functional programming alterations of multiple organs in offspring induced by medication during pregnancy, which can provide a theoretical and experimental basis for rational medication during pregnancy and effective prevention and treatment of drug-related multiple fetal-originated diseases.
Collapse
Affiliation(s)
- Zhengjie Lu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Yongguo Dai
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Science, Wuhan 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disorder, Wuhan 430071, China
| |
Collapse
|
2
|
Goetzl L, Darbinian N, Merabova N, Devane LC, Ramamoorthy S. Gestational Age Variation in Human Placental Drug Transporters. Front Pharmacol 2022; 13:837694. [PMID: 35462922 PMCID: PMC9019509 DOI: 10.3389/fphar.2022.837694] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/24/2022] [Indexed: 12/29/2022] Open
Abstract
Patient and providers’ fear of fetal exposure to medications may lead to discontinuation of treatment, disease relapse, and maternal morbidity. Placental drug transporters play a critical role in fetal exposure through active transport but the majority of data are limited to the 3rd trimester, when the majority of organogenesis has already occurred. Our objective was to define gestational age (GA) dependent changes in protein activity, expression and modifications of five major placental drug transporters: SERT, P-gp, NET, BCRP and MRP3. Apical brush border membrane fractions were prepared from fresh 1st, 2nd and 3rd trimester human placentas collected following elective pregnancy termination or planned cesarean delivery. A structured maternal questionnaire was used to identify maternal drug use and exclude exposed subjects. Changes in placental transporter activity and expression relative to housekeeping proteins were quantified. There was evidence for strong developmental regulation of SERT, NET, P-gp, BCRP and MRP3. P-gp and BCRP decreased with gestation (r = −0.72, p < 0.001 and r = −0.77, p < 0.001, respectively). Total SERT increased with gestation but this increase was due to a decrease in SERT cleavage products across trimesters. Uncleaved SERT increased with GA (r = 0.89, p < 0.001) while cleaved SERT decreased with GA (r = −0.94, p < 0.001). Apical membrane NET overall did not appear to be developmentally regulated (r = −0.08, p = 0.53). Two forms of MRP3 were identified; the 50 kD form did not change across GA; the 160 kD form was steady in the 1st and 2nd trimester and increased in the 3rd trimester (r = 0.24, p = 0.02). The 50 kD form was expressed at higher levels. The observed patterns of SERT, NET P-gp, BCRP and MRP3 expression and activity may be associated with transporter activity or decreased placental permeability in the 1st trimester to transporter specific substrates including commonly used psychoactive medications such as anti-depressants, anti-psychotics, and amphetamines, while transport of nutrients and serotonin is important in the 1st trimester. Overall these observations are consistent with a strong protective effect during organogenesis. 3rd trimester estimates of fetal exposure obtained from cord blood likely significantly overestimate early fetal exposure to these medications at any fixed maternal dose.
Collapse
|
3
|
Fokina VM, Patrikeeva S, Wang XM, Noguchi S, Tomi M, König J, Ahmed MS, Nanovskaya T. Role of Uptake Transporters OAT4, OATP2A1, and OATP1A2 in Human Placental Bio-disposition of Pravastatin. J Pharm Sci 2022; 111:505-516. [PMID: 34597623 PMCID: PMC8792198 DOI: 10.1016/j.xphs.2021.09.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 02/03/2023]
Abstract
Pravastatin is currently under evaluation for prevention of preeclampsia. Factors contributing to placental disposition of pravastatin are important in assessment of potential undesirable fetal effects. The purpose of this study was to identify the uptake transporters that contribute to the placental disposition of pravastatin. Our data revealed the expression of organic anion transporting polypeptide 1A2 (OATP1A2) and OATP2A1 in the apical, and OATP2B1 and OATP5A1 in the basolateral membranes of the placenta, while organic anion transporter 4 (OAT4) exhibited higher expression in basolateral membrane but was detected in both membranes. Preloading placental membrane vesicles with glutarate increased the uptake of pravastatin suggesting involvement of glutarate-dependent transporters such as OAT4. In the HEK293 cells overexpressing individual uptake transporters, OATP2A1, OATP1A2 and OAT4 were determined to accept pravastatin as a substrate at physiological pH, while the uptake of pravastatin by OATP2B1 (known to interact with pravastatin at acidic pH) and OATP5A1 was not detected at pH 7.4. These findings led us to propose that OATP1A2 and OATP2A1 are responsible for the placental uptake of pravastatin from the maternal circulation, while OAT4 mediates the passage of the drug across placental basolateral membrane in the fetal-to-maternal direction.
Collapse
Affiliation(s)
- Valentina M Fokina
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Svetlana Patrikeeva
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiao-ming Wang
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Saki Noguchi
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, 105-8512, Japan
| | - Masatoshi Tomi
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo, 105-8512, Japan
| | - Jörg König
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054 Erlangen
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tatiana Nanovskaya
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
4
|
Fokina V, Patrikeeva S, Wang X, Shah M, Shah P, Russell WK, Ahmed MS, Rytting E, Nanovskaya T. Physicochemical and Biological Properties of Membrane Vesicles Derived from Human Term Placentas. J Biomed Nanotechnol 2022; 18:589-599. [PMID: 35484740 DOI: 10.1166/jbn.2022.3255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The purpose of this study was to conduct initial characterization of membrane vesicles isolated from human placenta by agitation of villous tissue (apical and basal) as well as vesicles obtained following dual perfusion of placental lobule. The morphology, physical and biological properties of the isolated vesicles were determined by electron microscopy, dynamic light scattering, and immunoblotting as well as nanoflow liquid chromatography-mass spectrometry proteomics analysis. CD-1 male mice were used to test the biocompatibility of the vesicles in vivo and assess the biodistribution of fluorescently labeled apical and perfusion vesicles. The vesicles obtained following placental perfusion and the apical vesicles had Z-average diameters of 199±23 nm and 246±24 nm, respectively, and demonstrated nanocarrier stability, low toxicity, and low immunogenicity. On the other hand, administration of basal vesicles resulted in animal demise with LD50 of 0.85 μgprotein/g. Both fluorescently labeled apical and perfusion vesicles were detected in the lungs, liver, kidneys, and spleen of CD-1 mice within 24 h of administration. However, there were differences in organ distribution of these vesicles over 24 hours time period. These data suggest that placental apical and perfusion vesicles have a potential for further development as biological vehicles for drug delivery.
Collapse
Affiliation(s)
- Valentina Fokina
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Svetlana Patrikeeva
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xiaoming Wang
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mansi Shah
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Poonam Shah
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Erik Rytting
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tatiana Nanovskaya
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
5
|
Ganesan M, Kanimozhi G, Pradhapsingh B, Khan HA, Alhomida AS, Ekhzaimy A, Brindha GR, Prasad NR. Phytochemicals reverse P-glycoprotein mediated multidrug resistance via signal transduction pathways. Biomed Pharmacother 2021; 139:111632. [PMID: 34243600 DOI: 10.1016/j.biopha.2021.111632] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023] Open
Abstract
P-glycoprotein, encoded by ATP-binding cassette transporters B1 gene (ABCB1), renders multidrug resistance (MDR) during cancer chemotherapy. Several synthetic small molecule inhibitors affect P-glycoprotein (P-gp) transport function in MDR tumor cells. However, inhibition of P-gp transport function adversely accumulates chemotherapeutic drugs in non-target normal tissues. Moreover, most small-molecule P-gp inhibitors failed in the clinical trials due to the low therapeutic window at the maximum tolerated dose. Therefore, downregulation of ABCB1-gene expression (P-gp) in tumor tissues seems to be a novel approach rather than inhibiting its transport function for the reversal of multidrug resistance (MDR). Several plant-derived phytochemicals modulate various signal transduction pathways and inhibit translocation of transcription factors, thereby reverses P-gp mediated MDR in tumor cells. Therefore, phytochemicals may be considered an alternative to synthetic small molecule P-gp inhibitors for the reversal of MDR in cancer cells. This review discussed the role of natural phytochemicals that modulate ABCB1 expression through various signal transduction pathways in MDR cancer cells. Therefore, modulating the cell signaling pathways by phytochemicals might play crucial roles in modulating ABCB1 gene expression and the reversal of MDR.
Collapse
Affiliation(s)
- M Ganesan
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - G Kanimozhi
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, Tamil Nadu, India
| | - B Pradhapsingh
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India
| | - Haseeb A Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhomida
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Aishah Ekhzaimy
- Division of Endocrinology, Department of Medicine, King Khalid University Hospital, Riyadh 12372, Saudi Arabia
| | - G R Brindha
- School of Computing, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamil Nadu, India
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar 608002, Tamil Nadu, India.
| |
Collapse
|
6
|
Vatter T, Klumpp L, Ganser K, Stransky N, Zips D, Eckert F, Huber SM. Against Repurposing Methadone for Glioblastoma Therapy. Biomolecules 2020; 10:biom10060917. [PMID: 32560384 PMCID: PMC7356722 DOI: 10.3390/biom10060917] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Methadone, which is used as maintenance medication for outpatient treatment of opioid dependence or as an analgesic drug, has been suggested by preclinical in vitro and mouse studies to induce cell death and sensitivity to chemo- or radiotherapy in leukemia, glioblastoma, and carcinoma cells. These data together with episodical public reports on long-term surviving cancer patients who use methadone led to a hype of methadone as an anti-cancer drug in social and public media. However, clinical evidence for a tumoricidal effect of methadone is missing and prospective clinical trials, except in colorectal cancer, are not envisaged because of the limited preclinical data available. The present article reviews the pharmacokinetics, potential molecular targets, as well as the evidence for a tumoricidal effect of methadone in view of the therapeutically achievable doses in the brain. Moreover, it provides original in vitro data showing that methadone at clinically relevant concentrations fails to impair clonogenicity or radioresistance of glioblastoma cells.
Collapse
Affiliation(s)
- Tatjana Vatter
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Lukas Klumpp
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
| | - Nicolai Stransky
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, 72076 Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University of Tübingen, 72076 Tübingen, Germany; (T.V.); (L.K.); (K.G.); (N.S.); (D.Z.); (F.E.)
- Correspondence: ; Tel.: +49-(0)7071-29-82183
| |
Collapse
|
7
|
Parikh A, Gopalakrishnan M, Azeem A, Booth A, El-Metwally D. Racial association and pharmacotherapy in neonatal opioid withdrawal syndrome. J Perinatol 2019; 39:1370-1376. [PMID: 31388115 DOI: 10.1038/s41372-019-0440-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/15/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine if racial differences are associated with Neonatal Opioid Withdrawal Syndrome (NOWS) severity. STUDY DESIGN A 10-year (2008-2017) retrospective cohort of infants ≥35 weeks gestation with prenatal exposure to opioids was included. The primary measure was the need for pharmacotherapy. Multivariable logistic regression and propensity score analysis were performed. RESULTS Among 345 infants with NOWS, 111 (32%) were black infants with 70% of them requiring pharmacotherapy as compared with 84% of white infants. Upon adjusting for significant covariates (methadone, benzodiazepine use, and gestational age), black infants were 57% less likely than whites to require pharmacotherapy (Odds ratio: 0.43, 95%CI: 0.22-0.80, p = 0.009). Similar results were observed with propensity score analysis. CONCLUSIONS Significant racial disparity observed may be secondary to genetic variations in opioid pharmacogenomics and/or extrinsic factors. Large-scale studies are warranted to include race in predictive models for early pharmacological intervention.
Collapse
Affiliation(s)
- Abhinav Parikh
- University of Maryland Medical Center, Baltimore, MD, USA.
| | - Mathangi Gopalakrishnan
- Center for Translational Medicine, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Ahad Azeem
- Long Island Jewish Forrest Hill Hospital, Forrest Hill, NY, 11375, USA
| | | | | |
Collapse
|
8
|
Marei HE, Casalbore P, Althani A, Coccè V, Cenciarelli C, Alessandri G, Brini AT, Parati E, Bondiolotti G, Pessina A. Human Olfactory Bulb Neural Stem Cells (Hu-OBNSCs) Can Be Loaded with Paclitaxel and Used to Inhibit Glioblastoma Cell Growth. Pharmaceutics 2019; 11:45. [PMID: 30669623 PMCID: PMC6358986 DOI: 10.3390/pharmaceutics11010045] [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: 12/07/2018] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Exploitation of the potential ability of human olfactory bulb (hOB) cells to carry, release, and deliver an effective, targeted anticancer therapy within the central nervous system (CNS) milieu remains elusive. Previous studies have demonstrated the marked ability of several types of stem cells (such as mesenchymal stem cells (MSCs) to carry and release different anti-cancer agents such as paclitaxel (PTX). Herein we investigate the ability of human olfactory bulb neural stem cells (Hu-OBNSCs) to carry and release paclitaxel, producing effective cytotoxic effects against cancer cells. We isolated Hu-OBNSCs from the hOB, uploaded them with PTX, and studied their potential cytotoxic effects against cancer cells in vitro. Interestingly, the Hu-OBNSCs displayed a five-fold increase in their resistance to the cytotoxicity of PTX, and the PTX-uploaded Hu-OBNSCs were able to inhibit proliferation and invasion, and to trigger marked cytotoxic effects on glioblastoma multiforme (GBM) cancer cells, and Human Caucasian fetal pancreatic adenocarcinoma 1 (CFPAC-1) in vitro. Despite their ability to resist the cytotoxic activity of PTX, the mechanism by which Hu-OBNSCs acquire resistance to PTX is not yet explained. Collectively our data indicate the ability of the Hu-OBNSCs to resist PTX, and to trigger effective cytotoxic effects against GBM cancer cells and CFPAC-1. This indicates their potential to be used as a carrier/vehicle for targeted anti-cancer therapy within the CNS.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Patrizia Casalbore
- Institute of Cell Biology and Neurobiology, National Research Council of Italy, 00015 Rome, Italy.
| | - Asmaa Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Valentina Coccè
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council of Italy, 00133 Rome, Italy.
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, 20133 Milan, Italy.
| | - Anna T Brini
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy.
| | - Eugenio Parati
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, IRCCS Neurological Institute C. Besta, 20133 Milan, Italy.
| | - Gianpietro Bondiolotti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20129 Milan, Italy.
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
9
|
Afrouzian M, Al-Lahham R, Patrikeeva S, Xu M, Fokina V, Fischer WG, Abdel-Rahman SZ, Costantine M, Ahmed MS, Nanovskaya T. Role of the efflux transporters BCRP and MRP1 in human placental bio-disposition of pravastatin. Biochem Pharmacol 2018; 156:467-478. [PMID: 30217571 DOI: 10.1016/j.bcp.2018.09.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
The expression and activity of human placental transporters during pregnancy could be altered by several factors including pathological changes associated with preeclampsia. The aims of this study were to identify the placental efflux transporters involved in the bio-disposition of pravastatin, determine the protein expression of these transporters and their encoding genes as well as the activity of pravastatin uptake in placentas obtained from patients with preeclampsia. ATP-dependent uptake of [3H]-pravastatin by trophoblast tissue apical and basal membrane vesicles exhibited sigmoidal kinetics. The curved shapes of Eadie-Hofstee plots indicate that more than one placental transporter are involved in the uptake of pravastatin. ATP-dependent uptake of [3H]-pravastatin into vesicles expressing MRP1-5, BCRP, and P-gp, as well as the results of inhibition studies suggest that BCRP and MRP1 are the major placental efflux transporters responsible for the in vitro uptake of pravastatin. Compared to placentas from healthy pregnancies, preeclamptic placentas had increased number of syncytial knots with increased expression of BCRP in their apical membrane and increased expression of MRP1 in the cytoplasm of the syncytiotrophoblast and in cytoplasm of syncytial knots. There was a concomitant increase in ABCC1 but not in ABCG2 gene expressions in preeclamptic placentas. ATP-dependent uptake of [3H]-pravastatin by vesicles prepared from apical membranes of preeclamptic placentas was similar to the uptake by vesicles prepared from placentas obtained after uncomplicated pregnancies (13.9 ± 6.5 vs 14.1 ± 5.8 pmol·mg protein-1 min-1). The transporter-specific changes in the expression of BCRP and MRP1 in preeclamptic placentas did not affect the efflux activity of transporters localized on the apical membrane of the syncytiotrophoblast.
Collapse
Affiliation(s)
- Marjan Afrouzian
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rabab Al-Lahham
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Svetlana Patrikeeva
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Meixiang Xu
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Valentina Fokina
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Wayne G Fischer
- Department of Qulity Management & Patient Safety, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sherif Z Abdel-Rahman
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Maged Costantine
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mahmoud S Ahmed
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Tatiana Nanovskaya
- Maternal-Fetal Pharmacology and Bio-Development Laboratories, Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
10
|
Al-Enazy S, Ali S, Albekairi N, El-Tawil M, Rytting E. Placental control of drug delivery. Adv Drug Deliv Rev 2017; 116:63-72. [PMID: 27527665 DOI: 10.1016/j.addr.2016.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/25/2016] [Accepted: 08/04/2016] [Indexed: 01/04/2023]
Abstract
The placenta serves as the interface between the maternal and fetal circulations and regulates the transfer of oxygen, nutrients, and waste products. When exogenous substances are present in the maternal bloodstream-whether from environmental contact, occupational exposure, medication, or drug abuse-the extent to which this exposure affects the fetus is determined by transport and biotransformation processes in the placental barrier. Advances in drug delivery strategies are expected to improve the treatment of maternal and fetal diseases encountered during pregnancy.
Collapse
|
11
|
Joshi AA, Vaidya SS, St-Pierre MV, Mikheev AM, Desino KE, Nyandege AN, Audus KL, Unadkat JD, Gerk PM. Placental ABC Transporters: Biological Impact and Pharmaceutical Significance. Pharm Res 2016; 33:2847-2878. [PMID: 27644937 DOI: 10.1007/s11095-016-2028-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/23/2016] [Indexed: 01/02/2023]
Abstract
The human placenta fulfills a variety of essential functions during prenatal life. Several ABC transporters are expressed in the human placenta, where they play a role in the transport of endogenous compounds and may protect the fetus from exogenous compounds such as therapeutic agents, drugs of abuse, and other xenobiotics. To date, considerable progress has been made toward understanding ABC transporters in the placenta. Recent studies on the expression and functional activities are discussed. This review discusses the placental expression and functional roles of several members of ABC transporter subfamilies B, C, and G including MDR1/P-glycoprotein, the MRPs, and BCRP, respectively. Since placental ABC transporters modulate fetal exposure to various compounds, an understanding of their functional and regulatory mechanisms will lead to more optimal medication use when necessary in pregnancy.
Collapse
Affiliation(s)
- Anand A Joshi
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, 23298-0533, USA
| | - Soniya S Vaidya
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, 23298-0533, USA
- Novartis Institutes of Biomedical Research, Cambridge, Massachusetts, USA
| | - Marie V St-Pierre
- Department of Clinical Pharmacology and Toxicology, University of Zurich Hospital, Zurich, Switzerland
| | - Andrei M Mikheev
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, Washington, USA
- Department of Neurosurgery, Institute of Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, Washington, 98109, USA
| | - Kelly E Desino
- Department of Pharmaceutical Chemistry, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
- Abbvie Inc, North Chicago, Illinois, USA
| | - Abner N Nyandege
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, 23298-0533, USA
| | - Kenneth L Audus
- Department of Pharmaceutical Chemistry, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington School of Pharmacy, Seattle, Washington, USA
| | - Phillip M Gerk
- Department of Pharmaceutics, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, 23298-0533, USA.
| |
Collapse
|
12
|
Transport of digoxin-loaded polymeric nanoparticles across BeWo cells, an in vitro model of human placental trophoblast. Ther Deliv 2016; 6:1325-34. [PMID: 26652279 DOI: 10.4155/tde.15.79] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Fetal arrhythmias can lead to fetal congestive heart failure and hydrops fetalis. Digoxin (the first-line treatment) has low transplacental permeability and high risk of maternal side effects. Biodegradable digoxin-loaded PEGylated poly(lactic-co-glycolic acid) nanoparticles may increase digoxin transport across BeWo b30 cell monolayers (an in vitro model of trophoblast in human placenta) by reducing the drug's interaction with P-gp. Results/methodology: The nanoparticles showed high encapsulation efficiency and sustained release over 48 h. Transport studies revealed significantly increased permeability across BeWo cell layers of digoxin-loaded nanoparticles when compared with free digoxin. P-gp inhibition also increased the permeability of digoxin, but not digoxin-loaded nanoparticles. CONCLUSION This represents a novel treatment strategy for fetal cardiovascular disease which may improve maternal and fetal outcomes.
Collapse
|
13
|
Daud ANA, Bergman JEH, Bakker MK, Wang H, Kerstjens-Frederikse WS, de Walle HEK, Groen H, Bos JHJ, Hak E, Wilffert B. P-Glycoprotein-Mediated Drug Interactions in Pregnancy and Changes in the Risk of Congenital Anomalies: A Case-Reference Study. Drug Saf 2016; 38:651-9. [PMID: 26017034 PMCID: PMC4486783 DOI: 10.1007/s40264-015-0299-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Introduction Drug use in pregnancy is very common but may cause harm to the fetus. The teratogenic effect of a drug is partly dependent on the drug level in the fetal circulation, which is associated with the transport across the placenta. Many drugs are substrates of P-glycoprotein (P-gp), an efflux transporter that acts as a protective barrier for the fetus. We aim to identify whether drug interactions associated with P-gp promote any changes in fetal drug exposure, as measured by the risk of having children with congenital anomalies. Methods In this study, cases (N = 4634) were mothers of children with congenital anomalies registered in the EUROCAT Northern Netherlands registry, and the reference population were mothers of children (N = 25,126) from a drug prescription database (IADB.nl). Results Drugs that are associated with P-gp transport were commonly used in pregnancy in cases (10 %) and population (12 %). Several drug classes, which are substrates for P-gp, were shown to have a higher user rate in mothers of cases with specific anomalies. The use of this subset of drugs in combination with other P-gp substrates increased the risk for specific anomalies (odds ratio [OR] 4.17, 95 % CI 1.75–9.91), and the addition of inhibitors further increased the risk (OR 13.03, 95 % CI 3.37–50.42). The same pattern of risk increment was observed when the drugs were analyzed separately according to substrate specificity. Conclusions The use of drugs associated with P-gp transport was common during pregnancy. For several drug classes associated with specific anomalies, P-gp-mediated drug interactions are associated with an increased risk for those specific anomalies. Electronic supplementary material The online version of this article (doi:10.1007/s40264-015-0299-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aizati N A Daud
- Unit of Pharmacotherapy and Pharmaceutical Care, Department of Pharmacy, University of Groningen, Groningen, The Netherlands,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bonomi A, Silini A, Vertua E, Signoroni PB, Coccè V, Cavicchini L, Sisto F, Alessandri G, Pessina A, Parolini O. Human amniotic mesenchymal stromal cells (hAMSCs) as potential vehicles for drug delivery in cancer therapy: an in vitro study. Stem Cell Res Ther 2015; 6:155. [PMID: 26315881 PMCID: PMC4552458 DOI: 10.1186/s13287-015-0140-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 07/29/2015] [Accepted: 07/29/2015] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION In the context of drug delivery, mesenchymal stromal cells (MSCs) from bone marrow and adipose tissue have emerged as interesting candidates due to their homing abilities and capacity to carry toxic loads, while at the same time being highly resistant to the toxic effects. Amongst the many sources of MSCs which have been identified, the human term placenta has attracted particular interest due to its unique, tissue-related characteristics, including its high cell yield and virtually absent expression of human leukocyte antigens and co-stimulatory molecules. Under basal, non-stimulatory conditions, placental MSCs also possess basic characteristics common to MSCs from other sources. These include the ability to secrete factors which promote cell growth and tissue repair, as well as immunomodulatory properties. The aim of this study was to investigate MSCs isolated from the amniotic membrane of human term placenta (hAMSCs) as candidates for drug delivery in vitro. METHODS We primed hAMSCs from seven different donors with paclitaxel (PTX) and investigated their ability to resist the cytotoxic effects of PTX, to upload the drug, and to release it over time. We then analyzed whether the uptake and release of PTX was sufficient to inhibit proliferation of CFPAC-1, a pancreatic tumor cell line sensitive to PTX. RESULTS For the first time, our study shows that hAMSCs are highly resistant to PTX and are not only able to uptake the drug, but also release it over time. Moreover, we show that PTX is released from hAMSCs in a sufficient amount to inhibit tumor cell proliferation, whilst some of the PTX is also retained within the cells. CONCLUSION Taken together, for the first time our results show that placental stem cells can be used as vehicles for the delivery of cytotoxic agents.
Collapse
Affiliation(s)
- Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| | - Antonietta Silini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Via Bissolati, 57 I-25124, Brescia, Italy.
| | - Elsa Vertua
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Via Bissolati, 57 I-25124, Brescia, Italy.
| | - Patrizia Bonassi Signoroni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Via Bissolati, 57 I-25124, Brescia, Italy.
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| | - Loredana Cavicchini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| | - Giulio Alessandri
- Cellular Neurobiology Laboratory, Department of Cerebrovascular Diseases, Fondazione IRCCS Neurological Institute C. Besta, Milan, Italy.
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy.
| | - Ornella Parolini
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Via Bissolati, 57 I-25124, Brescia, Italy.
| |
Collapse
|
15
|
Population approach to analyze the pharmacokinetics of free and total lopinavir in HIV-infected pregnant women and consequences for dose adjustment. Antimicrob Agents Chemother 2015; 59:5727-35. [PMID: 26149996 DOI: 10.1128/aac.00863-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/01/2015] [Indexed: 11/20/2022] Open
Abstract
The aims of this study were to describe the unbound and total lopinavir (LPV) pharmacokinetics in pregnant women in order to evaluate if a dosing adjustment is necessary during pregnancy. Lopinavir placental transfer is described, and several genetic covariates were tested to explain its variability. A total of 400 maternal, 79 cord blood, and 48 amniotic fluid samples were collected from 208 women for LPV concentration determinations and pharmacokinetics analysis. Among the maternal LPV concentrations, 79 samples were also used to measure the unbound LPV concentrations. Population pharmacokinetics models were developed by using NONMEM software. Two models were developed to describe (i) unbound and total LPV pharmacokinetics and (ii) LPV placental transfer. The pharmacokinetics was best described by a one-compartment model with first-order absorption and elimination. A pregnancy effect was found on maternal clearance (39% increase), whereas the treatment group (monotherapy versus triple therapy) or the genetic polymorphisms did not explain the pharmacokinetics or placental transfer of LPV. Efficient unbound LPV concentrations in nonpregnant women were similar to those measured during the third trimester of pregnancy. Our study showed a 39% increase of maternal total LPV clearance during pregnancy, whereas unbound LPV concentrations were similar to those simulated in nonpregnant women. The genetic polymorphisms selected did not influence the LPV pharmacokinetics or placental transfer. Thus, we suggest that the LPV dosage should not be increased during pregnancy.
Collapse
|
16
|
Gharavi R, Hedrich W, Wang H, Hassan HE. Transporter-Mediated Disposition of Opioids: Implications for Clinical Drug Interactions. Pharm Res 2015; 32:2477-502. [PMID: 25972096 DOI: 10.1007/s11095-015-1711-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/06/2015] [Indexed: 01/08/2023]
Abstract
Opioid-related deaths, abuse, and drug interactions are growing epidemic problems that have medical, social, and economic implications. Drug transporters play a major role in the disposition of many drugs, including opioids; hence they can modulate their pharmacokinetics, pharmacodynamics and their associated drug-drug interactions (DDIs). Our understanding of the interaction of transporters with many therapeutic agents is improving; however, investigating such interactions with opioids is progressing relatively slowly despite the alarming number of opioids-mediated DDIs that may be related to transporters. This review presents a comprehensive report of the current literature relating to opioids and their drug transporter interactions. Additionally, it highlights the emergence of transporters that are yet to be fully identified but may play prominent roles in the disposition of opioids, the growing interest in transporter genomics for opioids, and the potential implications of opioid-drug transporter interactions for cancer treatments. A better understanding of drug transporters interactions with opioids will provide greater insight into potential clinical DDIs and could help improve opioids safety and efficacy.
Collapse
Affiliation(s)
- Robert Gharavi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N Pine Street, Rooms: N525 (Office), Baltimore, Maryland, 21201, USA
| | | | | | | |
Collapse
|
17
|
Transplacental transfer and distribution of pravastatin. Am J Obstet Gynecol 2013; 209:373.e1-5. [PMID: 24070396 DOI: 10.1016/j.ajog.2013.05.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/02/2013] [Accepted: 05/13/2013] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The objective of the study was to determine the bidirectional transfer of pravastatin across the dually perfused term human placental lobule and its distribution between the tissue and maternal and fetal circuits. STUDY DESIGN The transfer of pravastatin was determined in the maternal-to-fetal (n = 11) and fetal-to-maternal (n = 10) directions. Pravastatin was coperfused with its [(3)H]-isotope and the marker compound antipyrine (20 μg/mL) and its [(14)C]-isotope. The concentration of pravastatin in the perfused tissue and the maternal and fetal circuits was determined using liquid scintillation spectrometry. Inside-out vesicles prepared from placental brush border membranes were utilized to investigate the role of efflux transporters in the transplacental transfer of pravastatin. RESULTS Pravastatin was transferred from the maternal to the fetal circuit and vice versa. In the maternal-to-fetal direction, the distribution of pravastatin at the end of experiment was as follows: 14 ± 5% of the drug was retained by the tissue, 68 ± 5% remained in the maternal circuit, and 18 ± 4% was transferred to the fetal circuit. The normalized transfer of pravastatin (clearance index) to antipyrine in the fetal-to-maternal direction (0.48 ± 0.07) was higher than its transfer in the maternal-to-fetal direction (0.36 ± 0.07, P < .01). Furthermore, pravastatin inhibited the adenosine triphosphate (ATP)-dependent uptake of the paclitaxel and estrone sulfate. CONCLUSION The transfer of pravastatin across the dually perfused placental lobule suggests that fetal exposure to pravastatin is plausible. The higher transfer of pravastatin in the fetal-to-maternal direction than the reverse as well as its inhibition of the ATP-dependent uptake of [(3)H]-paclitaxel and [(3)H]-estrone sulfate strongly suggest the involvement of efflux transporters in decreasing its transfer across the placenta and support pravastatin's favorable pharmacokinetic profile in pregnancy.
Collapse
|
18
|
Herédi-Szabó K, Palm JE, Andersson TB, Pál Á, Méhn D, Fekete Z, Beéry E, Jakab KT, Jani M, Krajcsi P. A P-gp vesicular transport inhibition assay – Optimization and validation for drug–drug interaction testing. Eur J Pharm Sci 2013; 49:773-81. [DOI: 10.1016/j.ejps.2013.04.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 02/18/2013] [Accepted: 04/30/2013] [Indexed: 12/16/2022]
|
19
|
Wang J, Cai B, Huang DX, Yang SD, Guo L. Decreased analgesic effect of morphine, but not buprenorphine, in patients with advanced P-glycoprotein(+) cancers. Pharmacol Rep 2013; 64:870-7. [PMID: 23087138 DOI: 10.1016/s1734-1140(12)70881-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 04/23/2012] [Indexed: 10/25/2022]
Abstract
BACKGROUND P-glycoprotein (P-gp) is expressed on the blood-brain barrier (BBB) and acts as a transporter regulating the analgesic effect of morphine. The P-gp is also expressed by different types of tumors. The aim of this study was to determine the potential association of the P-gp expression in malignant tumors with analgesic effects in patients. METHODS The P-gp expression in 120 malignant tumors was examined by immunohistochemistry. The analgesic responses of individual patients to morphine and buprenorphine (BNP) were evaluated by visual analog scale (VAS). The levels of plasma morphine and BNP were determined by HPLC. RESULTS We found that there was no significant difference in the values of VAS between patients with P-gp(+) and P-gp(-) malignant tumors in responses to 0.000025 g x kg(-2) of BNP administered by patient-controlled intravenous analgesia (PCIA), accompanied by similar levels of plasma BNP in those patients. In contrast, the values of VAS in response to 0.00075 g x kg(-2) of morphine in patients with P-gp(+) tumors were significantly greater than those in the patients with P-gp(-) tumors, although similar levels of plasma morphine were detected in both groups of patients. Furthermore, treatment with a higher dose (0.0011 g x kg(-2)) of morphine effectively controlled pain in those with P-gp(+) tumors. CONCLUSION Our data indicated that patients with P-gp(+) tumors required a higher dose of morphine to achieve an analgesic effect and that the P-gp expression in tumors may be valuable for predicting the analgesic responses of patients with severe pain to morphine.
Collapse
Affiliation(s)
- Jun Wang
- Department of Anesthesiology, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China.
| | | | | | | | | |
Collapse
|
20
|
Placental transfer of maraviroc in an ex vivo human cotyledon perfusion model and influence of ABC transporter expression. Antimicrob Agents Chemother 2013; 57:1415-20. [PMID: 23295922 DOI: 10.1128/aac.01821-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Nowadays, antiretroviral therapy is recommended during pregnancy to prevent mother-to-child transmission of HIV. However, for many antiretroviral drugs, including maraviroc, a CCR5 antagonist, very little data exist regarding placental transfer. Besides, various factors may modulate this transfer, including efflux transporters belonging to the ATP-binding cassette (ABC) transporter superfamily. We investigated maraviroc placental transfer and the influence of ABC transporter expression on this transfer using the human cotyledon perfusion model. Term placentas were perfused ex vivo for 90 min with maraviroc (600 ng/ml) either in the maternal-to-fetal (n = 10 placentas) or fetal-to-maternal (n = 6 placentas) direction. Plasma concentrations were determined by ultra performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS). Fetal transfer rates (FTR) and clearance indexes (CLI) were calculated as ratios of fetal to maternal concentrations at steady state (mean values between 30 and 90 min) and ratios of FTR of maraviroc to that of antipyrine, respectively. ABC transporter gene expression levels were determined by quantitative reverse transcription (RT)-PCR and ABCB1 protein expression by Western blotting. For the maternal-to-fetal direction, the mean FTR and CLI were 8.0% ± 3.0 and 0.26 ± 0.07, respectively, whereas the mean CLI was 0.52 ± 0.23 for the fetal-to-maternal direction. We showed a significant inverse correlation between maraviroc CLI and ABCC2, ABCC10, and ABCC11 placental gene expression levels (P < 0.05). To conclude, we report a low maraviroc placental transfer probably involving ABC efflux transporters and thus in all likelihood associated with a limited fetal exposition. Nevertheless, these results would need to be supported by in vivo data obtained from paired maternal and cord blood samples.
Collapse
|
21
|
Multidrug Resistance in Cancer: A Tale of ABC Drug Transporters. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2013. [DOI: 10.1007/978-1-4614-7070-0_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Rubinchik-Stern M, Eyal S. Drug Interactions at the Human Placenta: What is the Evidence? Front Pharmacol 2012; 3:126. [PMID: 22787449 PMCID: PMC3391695 DOI: 10.3389/fphar.2012.00126] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 06/14/2012] [Indexed: 01/16/2023] Open
Abstract
Pregnant women (and their fetuses) are treated with a significant number of prescription and non-prescription medications. Interactions among those drugs may affect their efficacy and toxicity in both mother and fetus. Whereas interactions that result in altered drug concentrations in maternal plasma are detectable, those involving modulation of placental transfer mechanisms are rarely reflected by altered drug concentrations in maternal plasma. Therefore, they are often overlooked. Placental-mediated interactions are possible because the placenta is not only a passive diffusional barrier, but also expresses a variety of influx and efflux transporters and drug-metabolizing enzymes. Current data on placental-mediated drug interactions are limited. In rodents, pharmacological or genetic manipulations of placental transporters significantly affect fetal drug exposure. In contrast, studies in human placentae suggest that the magnitude of such interactions is modest in most cases. Nevertheless, under certain circumstances, such interactions may be of clinical significance. This review describes currently known mechanisms of placental-mediated drug interactions and the potential implications of such interactions in humans. Better understanding of those mechanisms is important for minimizing fetal toxicity from drugs while improving their efficacy when directed to treat the fetus.
Collapse
|
23
|
Bartu AE, Ilett KF, Hackett LP, Doherty DA, Hamilton D. Buprenorphine exposure in infants of opioid-dependent mothers at birth. Aust N Z J Obstet Gynaecol 2012; 52:342-7. [PMID: 22428721 DOI: 10.1111/j.1479-828x.2012.01424.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 02/05/2012] [Indexed: 11/29/2022]
Affiliation(s)
- Anne E. Bartu
- Faculty of Health Sciences; School of Nursing and Midwifery; Curtin University of Technology; Australia
| | - Kenneth F. Ilett
- Pharmacology, Pharmacy and Anaesthesiology Unit; School of Medicine and Pharmacology; University of Western Australia; Australia
- Clinical Pharmacology and Toxicology Laboratory; Path West Laboratory Medicine; Australia
| | - L. Peter Hackett
- Clinical Pharmacology and Toxicology Laboratory; Path West Laboratory Medicine; Australia
| | - Dorota A. Doherty
- School of Women's and Infants' Health; University of Western Australia; Australia
| | - Dale Hamilton
- King Edward Memorial Hospital; Women and Newborn Health Services; Western Australia Australia
| |
Collapse
|
24
|
Cigarette Smoking and Neonatal Outcomes in Depressed and Non-Depressed Opioid-Dependent Agonist-Maintained Pregnant Patients. ADDICTIVE DISORDERS & THEIR TREATMENT 2011; 10:180-187. [PMID: 22833702 DOI: 10.1097/adt.0b013e31821cadbd] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
AIMS: To investigate whether cigarette smoking and/or depression contribute to neonatal abstinence syndrome (NAS) severity. DESIGN: Cohort study analyzing data from a randomized, controlled trial of methadone versus buprenorphine. SETTING: Seven study sites that randomized patients to study conditions and provided comprehensive addiction treatment to pregnant patients. PARTICIPANTS: 119 of 131 opioid-dependent pregnant patients who completed the MOTHER study. MEASUREMENTS: Smoking data and depression status were obtained from the Addiction Severity Index and Mini International Neuropsychiatric Interview, respectively. Neonatal outcomes (birth weight, preterm delivery and NAS pharmacologic treatment) were collected from the medical charts. Study site was a fixed-effect factor in all analyses. FINDINGS: Cigarette smoking was reported by 94% of participants and depression identified in 35%. Smoking was associated with low birth weight, preterm delivery, and NAS pharmacologic treatment in both depressed and non-depressed participants. The association between smoking and NAS treatment differed significantly between depressed and non-depressed participants. Among non-depressed participants, adjusting for site and illicit drug use, each additional average cigarette per day (CPD) increased the odds of NAS treatment by 12% [95%CI: (1.02-1.23), p=0.02]. Among depressed participants, each additional average CPD did not statistically increase the odds of NAS treatment [OR: 0.94, 95% CI: (0.84-1.04), p=0.23]. CONCLUSIONS: These results are consistent with the hypothesis that NAS expression is influenced by many factors. The relationship between CPD and NAS pharmacologic treatment is attenuated among depressed women in this study for reasons currently unknown. Further investigations are needed to clarify the complex relationships among maternal smoking, depression, and NAS.
Collapse
|
25
|
Wu CP, Ohnuma S, Ambudkar SV. Discovering natural product modulators to overcome multidrug resistance in cancer chemotherapy. Curr Pharm Biotechnol 2011; 12:609-20. [PMID: 21118092 DOI: 10.2174/138920111795163887] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 04/12/2010] [Indexed: 12/19/2022]
Abstract
Multidrug resistance caused by the overexpression of ABC drug transporters is a major obstacle in clinical cancer chemotherapy. For several years, it appeared that direct inhibition of ABC transporters would be the cheapest and most efficient way to combat this problem. Unfortunately, progress in finding a potent, selective inhibitor to modulate ABC transporters and restore drug sensitivity in multidrug-resistant cancer cells has been slow and challenging. Candidate drugs should ideally be selective, potent and relatively non-toxic. Many researchers in recent years have turned their attention to utilizing natural products as the building blocks for the development of the next generation of inhibitors, especially after the disappointing results obtained from inhibitors of the first three generations at the clinical trial stage. The first step is to discover natural substances (distinct from the first three generation inhibitors) that are potent, selective and relatively non-toxic in order to be used clinically. Here, we present a brief overview of the prospect of using natural products to modulate the function of ABC drug transporters clinically and their impact on human physiology and pharmacology.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
26
|
Ni Z, Mao Q. ATP-binding cassette efflux transporters in human placenta. Curr Pharm Biotechnol 2011; 12:674-85. [PMID: 21118087 DOI: 10.2174/138920111795164057] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 04/08/2010] [Indexed: 01/11/2023]
Abstract
Pregnant women are often complicated with diseases including viral or bacterial infections, epilepsy, hypertension, or pregnancy-induced conditions such as depression and gestational diabetes that require treatment with medication. In addition, substance abuse during pregnancy remains a major public health problem. Many drugs used by pregnant women are off label without the necessary dose, efficacy, and safety data required for rational dosing regimens of these drugs. Thus, a major concern arising from the widespread use of drugs by pregnant women is the transfer of drugs across the placental barrier, leading to potential toxicity to the developing fetus. Knowledge regarding the ATP-binding cassette (ABC) efflux transporters, which play an important role in drug transfer across the placental barrier, is absolutely critical for optimizing the therapeutic strategy to treat the mother while protecting the fetus during pregnancy. Such transporters include P-glycoprotein (P-gp, gene symbol ABCB1), the breast cancer resistance protein (BCRP, gene symbol ABCG2), and the multidrug resistance proteins (MRPs, gene symbol ABCCs). In this review, we summarize the current knowledge with respect to developmental expression and regulation, membrane localization, functional significance, and genetic polymorphisms of these ABC transporters in the placenta and their relevance to fetal drug exposure and toxicity.
Collapse
Affiliation(s)
- Zhanglin Ni
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
27
|
Pollex EK, Hutson JR. Genetic polymorphisms in placental transporters: implications for fetal drug exposure to oral antidiabetic agents. Expert Opin Drug Metab Toxicol 2011; 7:325-39. [DOI: 10.1517/17425255.2011.553188] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
28
|
Jones HE, Kaltenbach K, Heil SH, Stine SM, Coyle MG, Arria AM, O'Grady KE, Selby P, Martin PR, Fischer G. Neonatal abstinence syndrome after methadone or buprenorphine exposure. N Engl J Med 2010; 363:2320-31. [PMID: 21142534 PMCID: PMC3073631 DOI: 10.1056/nejmoa1005359] [Citation(s) in RCA: 660] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Methadone, a full mu-opioid agonist, is the recommended treatment for opioid dependence during pregnancy. However, prenatal exposure to methadone is associated with a neonatal abstinence syndrome (NAS) characterized by central nervous system hyperirritability and autonomic nervous system dysfunction, which often requires medication and extended hospitalization. Buprenorphine, a partial mu-opioid agonist, is an alternative treatment for opioid dependence but has not been extensively studied in pregnancy. METHODS We conducted a double-blind, double-dummy, flexible-dosing, randomized, controlled study in which buprenorphine and methadone were compared for use in the comprehensive care of 175 pregnant women with opioid dependency at eight international sites. Primary outcomes were the number of neonates requiring treatment for NAS, the peak NAS score, the total amount of morphine needed to treat NAS, the length of the hospital stay for neonates, and neonatal head circumference. RESULTS Treatment was discontinued by 16 of the 89 women in the methadone group (18%) and 28 of the 86 women in the buprenorphine group (33%). A comparison of the 131 neonates whose mothers were followed to the end of pregnancy according to treatment group (with 58 exposed to buprenorphine and 73 exposed to methadone) showed that the former group required significantly less morphine (mean dose, 1.1 mg vs. 10.4 mg; P<0.0091), had a significantly shorter hospital stay (10.0 days vs. 17.5 days, P<0.0091), and had a significantly shorter duration of treatment for the neonatal abstinence syndrome (4.1 days vs. 9.9 days, P<0.003125) (P values calculated in accordance with prespecified thresholds for significance). There were no significant differences between groups in other primary or secondary outcomes or in the rates of maternal or neonatal adverse events. CONCLUSIONS These results are consistent with the use of buprenorphine as an acceptable treatment for opioid dependence in pregnant women. (Funded by the National Institute on Drug Abuse; ClinicalTrials.gov number, NCT00271219.).
Collapse
Affiliation(s)
- Hendrée E Jones
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hemauer SJ, Patrikeeva SL, Wang X, Abdelrahman DR, Hankins GDV, Ahmed MS, Nanovskaya TN. Role of transporter-mediated efflux in the placental biodisposition of bupropion and its metabolite, OH-bupropion. Biochem Pharmacol 2010; 80:1080-6. [PMID: 20599802 DOI: 10.1016/j.bcp.2010.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 06/10/2010] [Accepted: 06/14/2010] [Indexed: 10/19/2022]
Abstract
Cigarette smoking during pregnancy is a preventable risk factor associated with maternal and fetal complications. Bupropion is an antidepressant used successfully for smoking cessation in non-pregnant patients. Our goal is to determine whether it could benefit the pregnant patient seeking smoking cessation. The aim of this investigation was to determine the role of human placenta in the disposition of bupropion and its major hepatic metabolite, OH-bupropion. The expression of efflux transporters P-gp and BCRP was determined in placental brush border membrane (n=200) and revealed a positive correlation (p<0.05). Bupropion was transported by BCRP (K(t) 3 microM, V(max) 30 pmol/mg protein/min) and P-gp (K(t) 0.5 microM, V(max) 6 pmol/mg protein min) in placental inside-out vesicles (IOVs). OH-bupropion crossed the dually-perfused human placental lobule without undergoing further metabolism, nor was it an efflux substrate of P-gp or BCRP. In conclusion, our data indicate that human placenta actively regulates the disposition of bupropion (via metabolism, active transport), but not its major hepatic metabolite, OH-bupropion.
Collapse
Affiliation(s)
- Sarah J Hemauer
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0587, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Placental P-glycoprotein and breast cancer resistance protein: Influence of polymorphisms on fetal drug exposure and physiology. Placenta 2010; 31:351-7. [DOI: 10.1016/j.placenta.2010.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2010] [Revised: 02/13/2010] [Accepted: 02/16/2010] [Indexed: 12/11/2022]
|
31
|
Hemauer SJ, Patrikeeva SL, Nanovskaya TN, Hankins GDV, Ahmed MS. Role of human placental apical membrane transporters in the efflux of glyburide, rosiglitazone, and metformin. Am J Obstet Gynecol 2010; 202:383.e1-7. [PMID: 20350646 DOI: 10.1016/j.ajog.2010.01.035] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 12/22/2009] [Accepted: 01/15/2010] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Substrates of placental efflux transporters could compete for a single transporter, which could result in an increase in the transfer of each substrate to the fetal circulation. Our aim was to determine the role of placental transporters in the biodisposition of oral hypoglycemic drugs that could be used as monotherapy or in combination therapy for gestational diabetes. STUDY DESIGN Inside-out brush border membrane vesicles from term placentas were used to determine the efflux of glyburide, rosiglitazone, and metformin by P-glycoprotein, breast cancer resistance protein, and multidrug resistance protein. RESULTS Glyburide was transported by multidrug resistance protein (43 +/- 4%); breast cancer resistance protein (25 +/- 5%); and P-glycoprotein (9 +/- 5%). Rosiglitazone was transported predominantly by P-glycoprotein (71 +/- 26%). Metformin was transported by P-glycoprotein (58 +/- 20%) and breast cancer resistance protein (25 +/- 14%). CONCLUSION Multiple placental transporters contribute to efflux of glyburide, rosiglitazone, and metformin. Administration of drug combinations could lead to their competition for efflux transporters.
Collapse
Affiliation(s)
- Sarah J Hemauer
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | |
Collapse
|
32
|
Modulation of human placental P-glycoprotein expression and activity by MDR1 gene polymorphisms. Biochem Pharmacol 2009; 79:921-5. [PMID: 19896927 DOI: 10.1016/j.bcp.2009.10.026] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 10/29/2009] [Accepted: 10/29/2009] [Indexed: 11/23/2022]
Abstract
The ABC transporter P-glycoprotein is a product of the MDR1 gene and its function in human placenta is to extrude xenobiotics from the tissue thus decreasing fetal exposure. The goal of this investigation was to examine the effect of three polymorphisms in the MDR1 gene on the expression and activity of placental P-gp. In 199 term placentas examined, the C1236T variant was associated with 11% lower P-gp protein expression than wild-type, while the C3435T and G2677T/A variants each were associated with a 16% reduction (p<0.05). Homozygotes for the C1236T and C3435T variant allele (TT) were associated with 42% and 47% increase in placental P-gp transport activity, respectively (p=0.04 and p=0.02) of the prototypic substrate, [(3)H]-paclitaxel. These findings indicate that the C3435T and G2677T/A SNPs in MDR1 are significantly associated with decreased placental P-gp protein expression, while the C1236T and C3245T homozygous variants are significantly associated with an increase in its efflux activity.
Collapse
|