1
|
Lee WH, Sun CK, Chang CH, Yang MH, Tsai TH. The anti-COVID-19 drug Paxlovid crosses biological barriers of the placenta and brain in rats. NPJ VIRUSES 2024; 2:4. [PMID: 40295802 PMCID: PMC11702666 DOI: 10.1038/s44298-023-00013-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/10/2023] [Indexed: 04/30/2025]
Abstract
Paxlovid (nirmatrelvir plus ritonavir) is an orally available drug for the treatment of COVID-19 disease. However, limited information remains on the biological barrier transfer of nirmatrelvir. In the present study, we investigated whether nirmatrelvir crosses the blood-placenta barrier to reach the conceptus (the collective term for the fetus, placenta, and amniotic fluid) during pregnancy and the blood-brain barrier (BBB) in male rats. Additionally, in this study, gender and pregnancy issues were investigated. Multisite microdialysis coupled with validated UHPLC-MS/MS was developed to monitor nirmatrelvir levels in maternal blood and the conceptus in pregnant rats and of the blood and brain in male and nonpregnant female rats after administration of nirmatrelvir (15 mg/kg, i.v.) alone and nirmatrelvir (15 mg/kg, i.v.) combined with ritonavir (7 mg/kg, i.v.). Pharmacokinetic results showed that nirmatrelvir rapidly penetrates the blood-placenta barrier to reach the conceptus after administration of nirmatrelvir (15 mg/kg, i.v.) alone and nirmatrelvir (15 mg/kg, i.v.) combined with ritonavir (7 mg/kg, i.v.) in pregnant rats. Nirmatrelvir also crosses the BBB in male and nonpregnant female rats in the same dose regimen. Compared to sex and pregnancy factors, the results show that protein-unbound nirmatrelvir increased significantly during pregnancy and did not differ between nonpregnant female and male rats. The results indicated that the concentrations of nirmatrelvir in the blood, conceptus, and brain were higher than the effective concentration of 90% (total EC90 = 292 ng/mL, unbound EC90 = 90.5 ng/mL, 181 nM) after the administration of nirmatrelvir plus ritonavir. Ritonavir provides a synergistic pharmacokinetic effect. Pregnancy is an important issue with increased protein-unbound nirmatrelvir in the blood and tissues.
Collapse
Affiliation(s)
- Wan-Hsin Lee
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chung-Kai Sun
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Chun-Hao Chang
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 404, Taiwan.
- Department of Chemistry, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
2
|
Bourdin V, Bigot W, Vanjak A, Burlacu R, Lopes A, Champion K, Depond A, Amador-Borrero B, Sene D, Comarmond C, Mouly S. Drug-Drug Interactions Involving Dexamethasone in Clinical Practice: Myth or Reality? J Clin Med 2023; 12:7120. [PMID: 38002732 PMCID: PMC10672071 DOI: 10.3390/jcm12227120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/04/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Concomitant administration of multiple drugs frequently causes severe pharmacokinetic or pharmacodynamic drug-drug interactions (DDIs) resulting in the possibility of enhanced toxicity and/or treatment failure. The activity of cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), a drug efflux pump sharing localization and substrate affinities with CYP3A4, is a critical determinant of drug clearance, interindividual variability in drug disposition and clinical efficacy, and appears to be involved in the mechanism of numerous clinically relevant DDIs, including those involving dexamethasone. The recent increase in the use of high doses of dexamethasone during the COVID-19 pandemic have emphasized the need for better knowledge of the clinical significance of drug-drug interactions involving dexamethasone in the clinical setting. We therefore aimed to review the already published evidence for various DDIs involving dexamethasone in vitro in cell culture systems and in vivo in animal models and humans.
Collapse
Affiliation(s)
- Venceslas Bourdin
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - William Bigot
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Anthony Vanjak
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Ruxandra Burlacu
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Amanda Lopes
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Karine Champion
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Audrey Depond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Blanca Amador-Borrero
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
| | - Damien Sene
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Chloe Comarmond
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM U976, Hôpital Saint-Louis, 75010 Paris, France
| | - Stéphane Mouly
- Internal Medicine Department, Département Médico-Universitaire INVICTUS, Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris (APHP).Nord—Université Paris-Cité, 75010 Paris, France; (V.B.); (W.B.); (A.V.); (R.B.); (A.L.); (K.C.); (A.D.); (B.A.-B.); (D.S.); (C.C.)
- INSERM UMR-S1144, Hôpital Fernand Widal, 75010 Paris, France
| |
Collapse
|
3
|
Salerno SN, Capparelli EV, McIlleron H, Gerhart JG, Dumond JB, Kashuba AD, Denti P, Gonzalez D. Leveraging physiologically based pharmacokinetic modeling to optimize dosing for lopinavir/ritonavir with rifampin in pediatric patients. Pharmacotherapy 2023; 43:638-649. [PMID: 35607886 PMCID: PMC9684348 DOI: 10.1002/phar.2703] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/28/2022] [Indexed: 11/11/2022]
Abstract
STUDY OBJECTIVE Treatment of HIV and tuberculosis co-infection leads to significant mortality in pediatric patients, and treatment can be challenging due to the clinically significant drug-drug interaction (DDI) between lopinavir/ritonavir (LPV/RTV) and rifampin. Doubling LPV/RTV results in insufficient lopinavir trough concentrations in pediatric patients. The objective of this study was to leverage physiologically based pharmacokinetic (PBPK) modeling to optimize the adjusted doses of LPV/RTV in children receiving the WHO-revised doses of rifampin (15 mg/kg daily). DESIGN Adult and pediatric PBPK models for LPV/RTV with rifampin were developed, including CYP3A and P-glycoprotein inhibition and induction. SETTING (OR DATA SOURCE) Data for LPV/RTV model development and evaluation were available from the pediatric AIDS Clinical Trials Group. PATIENTS Dosing simulations were next performed to optimize dosing in children (2 months to 8 years of age). INTERVENTION Exposure following super-boosted LPV/RTV with 10 and 15 mg/kg PO daily rifampin was simulated. MEASUREMENTS AND MAIN RESULTS Simulated parameters were within twofold observations for LPV, RTV, and rifampin in adults and children ≥2 weeks old. The model predicted that, in healthy adults receiving 400/100 mg oral LPV/RTV twice daily (BID), co-treatment with 600 mg oral rifampin daily decreased the steady-state area under the concentration vs. time curve of LPV by 79%, in line with the observed change of 75%. Simulated and observed concentration profiles were comparable for LPV/RTV (230/57.5 mg/m2 ) PO BID without rifampin and 230/230 mg/m2 LPV/RTV PO BID with 10 mg/kg PO daily rifampin in pediatric patients. Sixteen mg/kg of super-boosted LPV (LPV/RTV 1:1) PO BID with 15 mg/kg PO daily rifampin achieved simulated LPV troughs >1 mg/L in ≥93% of virtual children weighing 3.0-24.9 kg, which was comparable with 10 mg/kg PO daily rifampin. CONCLUSIONS Super-boosted LPV/RTV with 15 mg/kg rifampin achieves therapeutic LPV troughs in HIV/TB-infected simulated children.
Collapse
Affiliation(s)
- Sara N. Salerno
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edmund V. Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Helen McIlleron
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Jacqueline G. Gerhart
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Julie B. Dumond
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela D.M. Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paolo Denti
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
4
|
Devine K, Villalobos E, Kyle CJ, Andrew R, Reynolds RM, Stimson RH, Nixon M, Walker BR. The ATP-binding cassette proteins ABCB1 and ABCC1 as modulators of glucocorticoid action. Nat Rev Endocrinol 2023; 19:112-124. [PMID: 36221036 DOI: 10.1038/s41574-022-00745-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 01/24/2023]
Abstract
Responses to hormones that act through nuclear receptors are controlled by modulating hormone concentrations not only in the circulation but also within target tissues. The role of enzymes that amplify or reduce local hormone concentrations is well established for glucocorticoid and other lipophilic hormones; moreover, transmembrane transporters have proven critical in determining tissue responses to thyroid hormones. However, there has been less consideration of the role of transmembrane transport for steroid hormones. ATP-binding cassette (ABC) proteins were first shown to influence the accumulation of glucocorticoids in cells almost three decades ago, but observations over the past 10 years suggest that differential transport propensities of both exogenous and endogenous glucocorticoids by ABCB1 and ABCC1 transporters provide a mechanism whereby different tissues are preferentially sensitive to different steroids. This Review summarizes this evidence and the new insights provided for the physiology and pharmacology of glucocorticoid action, including new approaches to glucocorticoid replacement.
Collapse
Affiliation(s)
- Kerri Devine
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Elisa Villalobos
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Catriona J Kyle
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ruth Andrew
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Rebecca M Reynolds
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark Nixon
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Musgrave K, Power K, Laffan M, O’Donnell JS, Thachil J, Maraveyas A. Practical Treatment Guidance for Cancer-Associated Thrombosis – Managing the Challenging Patient: A Consensus Statement. Crit Rev Oncol Hematol 2022; 171:103599. [PMID: 35065219 DOI: 10.1016/j.critrevonc.2022.103599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
|
6
|
Garcia-Varela L, Attia K, Sembrano JC, Jacquet O, Antunes IF, Kwizera C, Visser TJ, Dierckx RAJO, Elsinga PH, Luurtsema G. A new approach to produce [ 18F]MC225 via one-step synthesis, a PET radiotracer for measuring P-gp function. EJNMMI Radiopharm Chem 2021; 6:24. [PMID: 34264435 PMCID: PMC8282851 DOI: 10.1186/s41181-021-00139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/11/2021] [Indexed: 11/19/2022] Open
Abstract
Background [18F]MC225 is a radiotracer for imaging P-glycoprotein (P-gp) function at the blood-brain barrier. The P-gp function can be altered due to different factors, for instance, decreased P-gp function has been described in patients with Alzheimer’s or Parkinson’s Disease. The current applied radiosynthesis of [18F]MC225 involves 2 steps, including the distillation of the [18F] fluoroethylbromide intermediate. To develop a more robust synthetic procedure, it is of interest to produce the radiotracer via a 1-step synthesis. The present study describes a new synthetic approach to produce [18F]MC225 via direct 18F-fluorination. Moreover, we also provide the appropriate conditions for the automation of the synthesis. A mesylate precursor was synthesized via a multi-step synthetic route and used for the radiolabeling. The nucleophilic substitution of the mesylate group by [18F] Fluoride was automated in two different synthesis modules: IBA Synthera and Eckert and Ziegler PharmTracer (E&Z). Results The mesylate precursor was synthesized in 7 steps starting with 5-hydroxy-1-tetralone (commercially available) in practical yields. The stability of the precursor was improved via mesylate salt formation method. The radiolabeling was done by adding the mesylate precursor dissolved in DMF to the dried [18F]KF/K2.2.2 complex and heating at 140 °C for 30 min. Quality control by UPLC confirmed the production of [18F]MC225 with a molar activity (Am) higher than 100 GBq/micromole. The synthesis time in Synthera was 106 min and the product was obtained with a radiochemical purity higher than 95% and RCY of 6.5%, while the production in E&Z lasted 120 min and the product had a lower radiochemical purity (91%) and RCY (3.8%). Conclusions [18F]MC225 was successfully produced via a 1-step reaction. The procedure is suitable for automation using commercially available synthesis modules. The automation of the radiosynthesis in the Synthera module allows the production of the [18F]MC225 by a reliable and simple method.
Collapse
Affiliation(s)
- Lara Garcia-Varela
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Khaled Attia
- Symeres, Kadijk 3, 9747, AT, Groningen, the Netherlands
| | | | | | - Inês F Antunes
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Chantal Kwizera
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Ton J Visser
- Symeres, Kadijk 3, 9747, AT, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands.
| |
Collapse
|
7
|
Sarma A, Das MK. Nose to brain delivery of antiretroviral drugs in the treatment of neuroAIDS. MOLECULAR BIOMEDICINE 2020; 1:15. [PMID: 34765998 PMCID: PMC7725542 DOI: 10.1186/s43556-020-00019-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
NeuroAIDS (Neuro Acquired Immunodeficiency Syndrome) or HIV (Human Immunodeficiency Virus) associated neuronal abnormality is continuing to be a significant health issue among AIDS patients even under the treatment of combined antiretroviral therapy (cART). Injury and damage to neurons of the brain are the prime causes of neuroAIDS, which happens due to the ingress of HIV by direct permeation across the blood-brain barrier (BBB) or else via peripherally infected macrophage into the central nervous system (CNS). The BBB performs as a stringent barricade for the delivery of therapeutics drugs. The intranasal route of drug administration exhibits as a non-invasive technique to bypass the BBB for the delivery of antiretroviral drugs and other active pharmaceutical ingredients inside the brain and CNS. This method is fruitful for the drugs that are unable to invade the BBB to show its action in the CNS and thus erase the demand of systemic delivery and thereby shrink systemic side effects. Drug delivery from the nose to the brain/CNS takes very less time through both olfactory and trigeminal nerves. Intranasal delivery does not require the involvement of any receptor as it occurs by an extracellular route. Nose to brain delivery also involves nasal associated lymphatic tissues (NALT) and deep cervical lymph nodes. However, very little research has been done to explore the utility of nose to brain delivery of antiretroviral drugs in the treatment of neuroAIDS. This review focuses on the potential of nasal route for the effective delivery of antiretroviral nanoformulations directly from nose to the brain.
Collapse
Affiliation(s)
- Anupam Sarma
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India.,Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026 India
| | - Malay K Das
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam 786004 India
| |
Collapse
|
8
|
Marsousi N, Daali Y, Fontana P, Reny JL, Ancrenaz-Sirot V, Calmy A, Rudaz S, Desmeules JA, Samer CF. Impact of Boosted Antiretroviral Therapy on the Pharmacokinetics and Efficacy of Clopidogrel and Prasugrel Active Metabolites. Clin Pharmacokinet 2019; 57:1347-1354. [PMID: 29453687 DOI: 10.1007/s40262-018-0637-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES Prasugrel and clopidogrel are inhibitors of the ADP-P2Y12 platelet receptor used in acute coronary syndrome patients. They require bioactivation via isoenzymes such as cytochrome P450 (CYP) 3A4, CYP2C19 and CYP2B6. Ritonavir and cobicistat are potent CYP3A inhibitors, prescribed as pharmacokinetic (PK) enhancers in the treatment of human immunodeficiency virus (HIV) infection. METHODS In this study, the impact of boosted antiretroviral therapies (ARTs) on the PK of clopidogrel and prasugrel active metabolites (AMs), and on the efficacy of prasugrel and clopidogrel, were evaluated in a randomized crossover clinical trial. RESULTS A significantly lower exposure to clopidogrel AM [3.2-fold lower area under the concentration-time curve (AUC) and maximum plasma concentration (Cmax)] and prasugrel AM (2.1-fold and 1.7-fold lower AUC and Cmax) were demonstrated in HIV-infected patients treated with boosted ARTs compared with healthy controls; however, a differential impact was observed on platelet inhibition between clopidogrel and prasugrel. Clopidogrel 300 mg induced adequate (although modest) platelet inhibition in all healthy subjects, while platelet inhibition was insufficient in 44% of HIV patients. On the contrary, prasugrel 60 mg induced a potent platelet inhibition in both healthy and HIV-infected subjects. CONCLUSION Prasugrel appears to remain an adequate antiplatelet agent in HIV-infected patients and could be preferred to clopidogrel in this context, regardless of the metabolic interaction and inhibition of its bioactivation pathways.
Collapse
Affiliation(s)
- Niloufar Marsousi
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1211, Geneva, Switzerland
- School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Geneva, Switzerland
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1211, Geneva, Switzerland
- School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Geneva, Switzerland
- Swiss Center for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Pierre Fontana
- Faculty of Medicine, Geneva University, Geneva, Switzerland
- Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of General Internal Medicine, Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - Virginie Ancrenaz-Sirot
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1211, Geneva, Switzerland
| | - Alexandra Calmy
- Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Geneva, Switzerland
- Swiss Center for Applied Human Toxicology (SCAHT), Basel, Switzerland
| | - Jules Alexandre Desmeules
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1211, Geneva, Switzerland
- School of Pharmaceutical Sciences, Geneva and Lausanne Universities, Geneva, Switzerland
- Swiss Center for Applied Human Toxicology (SCAHT), Basel, Switzerland
- Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Caroline Flora Samer
- Division of Clinical Pharmacology and Toxicology, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1211, Geneva, Switzerland.
- Swiss Center for Applied Human Toxicology (SCAHT), Basel, Switzerland.
- Faculty of Medicine, Geneva University, Geneva, Switzerland.
| |
Collapse
|
9
|
Wang Y, Li Y, Shang D, Efferth T. Interactions between artemisinin derivatives and P-glycoprotein. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152998. [PMID: 31301971 DOI: 10.1016/j.phymed.2019.152998] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Artemisinin was isolated and identified in 1972, which was the starting point for a new era in antimalarial drug therapy. Furthermore, numerous studies have demonstrated that artemisinin and its derivatives exhibit considerable anticancer activity both in vitro, in vivo, and even in clinical Phase I/II trials. P-glycoprotein (P-gp) mediated multi-drug resistance (MDR) is one of the most serious causes of chemotherapy failure in cancer treatment. Interestingly, many artemisinin derivatives exhibit excellent ability to overcome P-gp mediated MDR and even show collateral sensitivity against MDR cancer cells. Furthermore, some artemisinin derivatives show P-gp-mediated MDR reversal activity. Therefore, the interaction between P-gp and artemisinin derivatives is important to develop novel combination treatment protocols with artemisinin derivatives and established anticancer drugs that are P-gp substrates. PURPOSE This systematic review provides an updated overview on the interaction between artemisinin derivatives and P-gp and the effect of artemisinin derivatives on the P-gp expression level. RESULTS Artemisinin derivatives exhibit multi-specific interactions with P-gp. The currently used artemisinin derivatives are not transported by P-gp. However, some of novel synthetized artemisinin derivatives exhibit P-gp substrate properties. Furthermore, many artemisinin derivatives act as P-gp inhibitors, which exhibit the potential to reverse MDR towards clinically used anticancer drugs. CONCLUSION Therefore, studies on the interaction between artemisinin derivatives and P-gp provide important information for the development of novel anti-cancer artemisinin derivatives to reverse P-gp mediated MDR and for the design of rational artemisinin-based combination therapies against cancer.
Collapse
Affiliation(s)
- Yulin Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yongjie Li
- Department of Chinese Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian China; College of Integrative Medicine, Dalian Medical University, Dalian, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy, Johannes Gutenberg University 55128 Mainz, Germany.
| |
Collapse
|
10
|
Dalpiaz A, Pavan B. Nose-to-Brain Delivery of Antiviral Drugs: A Way to Overcome Their Active Efflux? Pharmaceutics 2018; 10:pharmaceutics10020039. [PMID: 29587409 PMCID: PMC6027266 DOI: 10.3390/pharmaceutics10020039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 02/06/2023] Open
Abstract
Although several viruses can easily infect the central nervous system (CNS), antiviral drugs often show dramatic difficulties in penetrating the brain from the bloodstream since they are substrates of active efflux transporters (AETs). These transporters, located in the physiological barriers between blood and the CNS and in macrophage membranes, are able to recognize their substrates and actively efflux them into the bloodstream. The active transporters currently known to efflux antiviral drugs are P-glycoprotein (ABCB1 or P-gp or MDR1), multidrug resistance-associated proteins (ABCC1 or MRP1, ABCC4 or MRP4, ABCC5 or MRP5), and breast cancer resistance protein (ABCG2 or BCRP). Inhibitors of AETs may be considered, but their co-administration causes serious unwanted effects. Nasal administration of antiviral drugs is therefore proposed in order to overcome the aforementioned problems, but innovative devices, formulations (thermoreversible gels, polymeric micro- and nano-particles, solid lipid microparticles, nanoemulsions), absorption enhancers (chitosan, papaverine), and mucoadhesive agents (chitosan, polyvinilpyrrolidone) are required in order to selectively target the antiviral drugs and, possibly, the AET inhibitors in the CNS. Moreover, several prodrugs of antiretroviral agents can inhibit or elude the AET systems, appearing as interesting substrates for innovative nasal formulations able to target anti-Human Immunodeficiency Virus (HIV) agents into macrophages of the CNS, which are one of the most important HIV Sanctuaries of the body.
Collapse
Affiliation(s)
- Alessandro Dalpiaz
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121 Ferrara, Italy.
| | - Barbara Pavan
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
11
|
Effects of HIV-1 Tat and Methamphetamine on Blood-Brain Barrier Integrity and Function In Vitro. Antimicrob Agents Chemother 2017; 61:AAC.01307-17. [PMID: 28893794 DOI: 10.1128/aac.01307-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/05/2017] [Indexed: 12/28/2022] Open
Abstract
Human immunodeficiency (HIV) infection results in neurocognitive deficits in about one half of infected individuals. Despite systemic effectiveness, restricted antiretroviral penetration across the blood-brain barrier (BBB) is a major limitation in fighting central nervous system (CNS)-localized infection. Drug abuse exacerbates HIV-induced cognitive and pathological CNS changes. This study's purpose was to investigate the effects of the HIV-1 protein Tat and methamphetamine on factors affecting drug penetration across an in vitro BBB model. Factors affecting paracellular and transcellular flux in the presence of Tat and methamphetamine were examined. Transendothelial electrical resistance, ZO-1 expression, and lucifer yellow (a paracellular tracer) flux were aspects of paracellular processes that were examined. Additionally, effects on P-glycoprotein (P-gp) and multidrug resistance protein 1 (MRP-1) mRNA (via quantitative PCR [qPCR]) and protein (via immunoblotting) expression were measured; Pgp and MRP-1 are drug efflux proteins. Transporter function was examined after exposure of Tat with or without methamphetamine using the P-gp substrate rhodamine 123 and also using the dual P-gp/MRP-1 substrate and protease inhibitor atazanavir. Tat and methamphetamine elicit complex changes affecting transcellular and paracellular transport processes. Neither Tat nor methamphetamine significantly altered P-gp expression. However, Tat plus methamphetamine exposure significantly increased rhodamine 123 accumulation within brain endothelial cells, suggesting that treatment inhibited or impaired P-gp function. Intracellular accumulation of atazanavir was not significantly altered after Tat or methamphetamine exposure. Atazanavir accumulation was, however, significantly increased by simultaneous inhibition of P-gp and MRP. Collectively, our investigations indicate that Tat and methamphetamine alter aspects of BBB integrity without affecting net flux of paracellular compounds. Tat and methamphetamine may also affect several aspects of transcellular transport.
Collapse
|
12
|
Nduka SO, Okonta MJ, Ajaghaku DL, Ukwe CV. In vitro and in vivo cytochrome P450 3A enzyme inhibition by Aframomum melegueta and Denniettia tripetala extracts. ASIAN PAC J TROP MED 2017; 10:576-581. [DOI: 10.1016/j.apjtm.2017.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/15/2017] [Accepted: 04/20/2017] [Indexed: 10/19/2022] Open
|
13
|
Schaller SJ, Alam SM, Mao J, Zhao Y, Blobner M, Greenblatt DJ, Martyn JAJ. Pharmacokinetics cannot explain the increased effective dose requirement for morphine and midazolam in rats during their extended administration alone or in combination. J Pharm Pharmacol 2016; 69:82-88. [DOI: 10.1111/jphp.12663] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/16/2016] [Indexed: 11/28/2022]
Abstract
Abstract
Objectives
Chronic administration of morphine and midazolam, alone or in combination, can induce tolerance to their effects. Data showed that co-administration of morphine and midazolam increased effective dose requirement of morphine, exceeding that observed with morphine alone.
Methods
To elucidate the pharmacokinetic component to the tolerance, we administered midazolam (2 mg/kg) and morphine (10 mg/kg) alone or their combination daily to rats for 12 days followed by a pharmacokinetic study on day 13. On the study day, each animal received a single bolus dose of 5 mg/kg morphine, and 2 mg/kg of midazolam 30 s later. Multiple blood samples were obtained for 6 h. Plasma drug concentrations were assayed by mass spectrometry optimized for small samples.
Key findings
Mean morphine clearance was as follows: 22.2, 27.2, 26.0 and 23.4 l/h per kg in the saline–saline, saline–midazolam, saline–morphine and midazolam–morphine groups, respectively. Corresponding midazolam clearances were 32.8, 23.0, 22.2 and 31.1 l/h per kg. ANOVA indicated no significant differences among the four groups in the clearances, half-lives, and volumes of distribution. Morphine and midazolam clearances were significantly correlated (R2 = 0.48, P < 0.001).
Conclusions
This animal model suggests that altered pharmacokinetics cannot explain tolerance evidenced as increased dose requirement for morphine or midazolam, when administered alone or combination, for extended periods.
Collapse
Affiliation(s)
- Stefan J Schaller
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children – Boston, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Klinik fűr Anaesthesiologie, Klinikum rechts der Isar, Technische Universität Műnchen, Munich, Germany
| | - Saad M Alam
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children – Boston, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jianren Mao
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children – Boston, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yanli Zhao
- Program in Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA, USA
| | - Manfred Blobner
- Klinik fűr Anaesthesiologie, Klinikum rechts der Isar, Technische Universität Műnchen, Munich, Germany
| | - David J Greenblatt
- Program in Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA, USA
| | - J A Jeevendra Martyn
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children – Boston, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Alam C, Whyte-Allman SK, Omeragic A, Bendayan R. Role and modulation of drug transporters in HIV-1 therapy. Adv Drug Deliv Rev 2016; 103:121-143. [PMID: 27181050 DOI: 10.1016/j.addr.2016.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 12/15/2022]
Abstract
Current treatment of human immunodeficiency virus type-1 (HIV-1) infection involves a combination of antiretroviral drugs (ARVs) that target different stages of the HIV-1 life cycle. This strategy is commonly referred to as highly active antiretroviral therapy (HAART) or combined antiretroviral therapy (cART). Membrane-associated drug transporters expressed ubiquitously in mammalian systems play a crucial role in modulating ARV disposition during HIV-1 infection. Members of the ATP-binding cassette (ABC) and solute carrier (SLC) transporter superfamilies have been shown to interact with ARVs, including those that are used as part of first-line treatment regimens. As a result, the functional expression of drug transporters can influence the distribution of ARVs at specific sites of infection. In addition, pathological factors related to HIV-1 infection and/or ARV therapy itself can alter transporter expression and activity, thus further contributing to changes in ARV disposition and the effectiveness of HAART. This review summarizes current knowledge on the role of drug transporters in regulating ARV transport in the context of HIV-1 infection.
Collapse
Affiliation(s)
- Camille Alam
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 2S2, Canada
| | - Sana-Kay Whyte-Allman
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 2S2, Canada
| | - Amila Omeragic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 2S2, Canada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario M5S 2S2, Canada.
| |
Collapse
|
15
|
Silva R, Vilas-Boas V, Carmo H, Dinis-Oliveira RJ, Carvalho F, de Lourdes Bastos M, Remião F. Modulation of P-glycoprotein efflux pump: induction and activation as a therapeutic strategy. Pharmacol Ther 2015; 149:1-123. [PMID: 25435018 DOI: 10.1016/j.pharmthera.2014.11.013] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/03/2023]
Abstract
P-glycoprotein (P-gp) is an ATP-dependent efflux pump encoded by the MDR1 gene in humans, known to mediate multidrug resistance of neoplastic cells to cancer therapy. For several decades, P-gp inhibition has drawn many significant research efforts in an attempt to overcome this phenomenon. However, P-gp is also constitutively expressed in normal human epithelial tissues and, due to its broad substrate specificity, to its cellular polarized expression in many excretory and barrier tissues, and to its great efflux capacity, it can play a crucial role in limiting the absorption and distribution of harmful xenobiotics, by decreasing their intracellular accumulation. Such a defense mechanism can be of particular relevance at the intestinal level, by significantly reducing the intestinal absorption of the xenobiotic and, consequently, avoiding its access to the target organs. In this review, the current knowledge on this important efflux pump is summarized, and a new focus is brought on the therapeutic interest of inducing and/or activating P-gp for limiting the toxicity caused by its substrates. Several in vivo and in vitro studies validating the use of such a therapeutic strategy are discussed. An extensive literature search for reported P-gp inducers/activators and for the experimental models used in their characterization was conducted. Those studies demonstrate that effective antidotal pathways can be achieved by efficiently promoting the P-gp-mediated efflux of deleterious xenobiotics, resulting in a significant reduction in their intracellular levels and, consequently, in a significant reduction of their toxicity.
Collapse
Affiliation(s)
- Renata Silva
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Vânia Vilas-Boas
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Helena Carmo
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Ricardo Jorge Dinis-Oliveira
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; INFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, Advanced Institute of Health Sciences - North (ISCS-N), CESPU, CRL, Gandra, Portugal; Department of Legal Medicine and Forensic Sciences, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Félix Carvalho
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Biological Sciences Department, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
16
|
Alms D, Fedrowitz M, Römermann K, Noack A, Löscher W. Marked differences in the effect of antiepileptic and cytostatic drugs on the functionality of P-glycoprotein in human and rat brain capillary endothelial cell lines. Pharm Res 2014; 31:1588-604. [PMID: 24477677 DOI: 10.1007/s11095-013-1264-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 12/09/2013] [Indexed: 01/16/2023]
Abstract
PURPOSE The expression of P-glycoprotein (Pgp) is increased in brain capillary endothelial cells (BCECs) of patients with pharmacoresistant epilepsy. This may restrict the penetration of antiepileptic drugs (AEDs) into the brain. However, the mechanisms underlying increased Pgp expression in epilepsy patients are not known. One possibility is that AEDs induce the expression and functionality of Pgp in BCECs. Several older AEDs that induce human cytochrome P450 enzymes also induce Pgp in hepatocytes and enterocytes, but whether this extends to Pgp at the human BBB and to newer AEDs is not known. METHODS This prompted us to study the effects of various old and new AEDs on Pgp functionality in the human BCEC line, hCMEC/D3, using the rhodamine 123 (Rho123) efflux assay. For comparison, experiments were performed in two rat BCEC lines, RBE4 and GPNT, and primary cultures of rat and pig BCECs. Furthermore, known Pgp inducers, such as dexamethasone and several cytostatic drugs, were included in our experiments. RESULTS Under control conditions, GPNT cells exhibited the highest and RBE4 the lowest Pgp expression and Rho123 efflux, while intermediate values were determined in hCMEC/D3. Known Pgp inducers increased Rho123 efflux in all cell lines, but marked inter-cell line differences in effect size were observed. Of the various AEDs examined, only carbamazepine (100 μM) moderately increased Pgp functionality in hCMEC/D3, while valproate (300 μM) inhibited Pgp. CONCLUSIONS These data do not indicate that treatment with AEDs causes a clinically relevant induction in Pgp functionality in BCECs that form the BBB.
Collapse
Affiliation(s)
- Dana Alms
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | | | | | | | | |
Collapse
|
17
|
Karlsson L, Carlsson B, Hiemke C, Ahlner J, Bengtsson F, Schmitt U, Kugelberg FC. Altered brain concentrations of citalopram and escitalopram in P-glycoprotein deficient mice after acute and chronic treatment. Eur Neuropsychopharmacol 2013; 23:1636-44. [PMID: 23428338 DOI: 10.1016/j.euroneuro.2013.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 01/13/2013] [Accepted: 01/15/2013] [Indexed: 12/26/2022]
Abstract
According to both in vitro and in vivo data P-glycoprotein (P-gp) may restrict the uptake of several antidepressants into the brain, thus contributing to the poor success rate of current antidepressant therapies. The therapeutic activity of citalopram resides in the S-enantiomer, whereas the R-enantiomer is practically devoid of serotonin reuptake potency. To date, no in vivo data are available that address whether the enantiomers of citalopram and its metabolites are substrates of P-gp. P-gp knockout (abcb1ab (-/-)) and wild-type (abcb1ab (+/+)) mice underwent acute (single-dose) and chronic (two daily doses for 10 days) treatment with citalopram (10mg/kg) or escitalopram (5mg/kg) Serum and brain samples were collected 1-6h after the first or last i.p. injection for subsequent drug analysis by an enantioselective HPLC method. In brain, 3-fold higher concentrations of S- and R-citalopram, and its metabolites, were found in abcb1ab (-/-) mice than in abcb1ab (+/+) mice after both acute and chronic citalopram treatments. After escitalopram treatment, the S-citalopram brain concentration was 3-5 times higher in the knockout mice than in controls. The results provide novel evidence that the enantiomers of citalopram are substrates of P-gp. Possible clinical and toxicological implications of this finding need to be further elucidated.
Collapse
Affiliation(s)
- Louise Karlsson
- Department of Medical and Health Sciences, Division of Drug Research, Clinical Pharmacology, Linköping University, Linköping, Sweden
| | | | | | | | | | | | | |
Collapse
|
18
|
De Rosa MF, Robillard KR, Kim CJ, Hoque MT, Kandel G, Kovacs C, Kaul R, Bendayan R. Expression of membrane drug efflux transporters in the sigmoid colon of HIV-infected and uninfected men. J Clin Pharmacol 2013; 53:934-45. [PMID: 23856938 DOI: 10.1002/jcph.132] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 06/10/2013] [Indexed: 01/27/2023]
Abstract
The use of antiretroviral therapy (ART) as pre-exposure prophylaxis (PrEP) has gained global attention as a promising HIV prevention strategy in men who have sex with men. Permeability of these agents in the rectal mucosa may be partially regulated by interactions with drug efflux transporters, P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs) and/or breast cancer resistance protein (BCRP). The objective of this work was to investigate the expression of drug efflux transporters in recto-sigmoid colon tissues of HIV-infected and uninfected men, and evaluate the association of ART and/or HIV infection with drug transporter expression. MDR1/P-gp, MRPs (1-4) and BCRP mRNA and protein expression were detected in sigmoid colon biopsies of HIV-uninfected individuals. Biopsies from HIV-infected, ART-naïve participants revealed a significant downregulation of P-gp and MRP2 protein levels compared to HIV-uninfected individuals. Biopsies from HIV-infected ART-treated patients showed 1.9-fold higher P-gp protein expression and 1.5-fold higher MRP2 protein expression compared to the ones obtained from the HIV-infected ART-naïve patients. This is a first report demonstrating that HIV infection or ART could alter expression of drug efflux transporters in gut mucosa which in turn could affect the permeability of PrEP antiretroviral agents across this barrier, a highly vulnerable site of HIV transmission.
Collapse
|
19
|
Chan GNY, Saldivia V, Yang Y, Pang H, de Lannoy I, Bendayan R. In vivo induction of P-glycoprotein expression at the mouse blood-brain barrier: an intracerebral microdialysis study. J Neurochem 2013; 127:342-52. [PMID: 23777437 DOI: 10.1111/jnc.12344] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 06/10/2013] [Accepted: 06/13/2013] [Indexed: 11/27/2022]
Abstract
Intracerebral microdialysis was utilized to investigate the effect of P-glycoprotein (a drug efflux transporter) induction at the mouse blood-brain barrier (BBB) on brain extracellular fluid concentrations of quinidine, an established substrate of P-glycoprotein. Induction was achieved by treating male CD-1 mice for 3 days with 5 mg/kg/day dexamethasone (DEX), a ligand of the nuclear receptor, pregnane X receptor, and a P-glycoprotein inducer. Tandem liquid chromatography mass spectrometric method was used to quantify analytes in dialysate, blood and plasma. P-glycoprotein, pregnane X receptor and Cyp3a11 (metabolizing enzyme for quinidine) protein expression in capillaries and brain homogenates was measured by immunoblot analysis. Following quinidine i.v. administration, the average ratio of unbound quinidine concentrations in brain extracellular fluid (determined from dialysate samples) to plasma at steady state (375-495 min) or Kp, uu, ECF /Plasma in the DEX-treated animals was 2.5-fold lower compared with vehicle-treated animals. In DEX-treated animals, P-glycoprotein expression in brain capillaries was 1.5-fold higher compared with vehicle-treated animals while Cyp3a11 expression in brain capillaries was not significantly different between the two groups. These data demonstrate that P-gp induction mediated by DEX at the BBB can significantly reduce quinidine brain extracellular fluid concentrations by decreasing its brain permeability and further suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible. Applying microdialysis, distribution of quinidine, a P-gp substrate, in mouse brain extracellular fluid (ECF) was investigated following ligand-mediated P-glycoprotein (P-gp) induction at the blood-brain barrier (BBB). We demonstrated that a PXR agonist (dexamethasone) significantly up-regulated P-gp in brain capillaries and reduced quinidine brain ECF concentrations. Our data suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible.
Collapse
Affiliation(s)
- Gary N Y Chan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
20
|
Induction of P-glycoprotein by antiretroviral drugs in human brain microvessel endothelial cells. Antimicrob Agents Chemother 2013; 57:4481-8. [PMID: 23836171 DOI: 10.1128/aac.00486-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The membrane-associated drug transporter P-glycoprotein (P-gp) plays an essential role in drug efflux from the brain. Induction of this protein at the blood-brain barrier (BBB) could further affect the ability of a drug to enter the brain. At present, P-gp induction mediated by antiretroviral drugs at the BBB has not been fully investigated. Since P-gp expression is regulated by ligand-activated nuclear receptors, i.e., human pregnane X receptor (hPXR) and human constitutive androstane receptor (hCAR), these receptors could represent potential pathways involved in P-gp induction by antiretroviral drugs. The aims of this study were (i) to determine whether antiretroviral drugs currently used in HIV pharmacotherapy are ligands for hPXR or hCAR and (ii) to examine P-gp function and expression in human brain microvessel endothelial cells treated with antiretroviral drugs identified as ligands of hPXR and/or hCAR. Luciferase reporter gene assays were performed to examine the activation of hPXR and hCAR by antiretroviral drugs. The hCMEC/D3 cell line, which is known to display several morphological and biochemical properties of the BBB in humans, was used to examine P-gp induction following 72 h of exposure to these agents. Amprenavir, atazanavir, darunavir, efavirenz, ritonavir, and lopinavir were found to activate hPXR, whereas abacavir, efavirenz, and nevirapine were found to activate hCAR. P-gp expression and function were significantly induced in hCMEC/D3 cells treated with these drugs at clinical concentrations in plasma. Together, our data suggest that P-gp induction could occur at the BBB during chronic treatment with antiretroviral drugs identified as ligands of hPXR and/or hCAR.
Collapse
|
21
|
Rhee JS, Kim BM, Choi BS, Choi IY, Wu RSS, Nelson DR, Lee JS. Whole spectrum of cytochrome P450 genes and molecular responses to water-accommodated fractions exposure in the marine medaka. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2013; 47:4804-4812. [PMID: 23573833 DOI: 10.1021/es400186r] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Water-accommodated fractions (WAFs) of crude oil include chemicals that are potent toxicants in fish. Increasing oil pollution thus demands a better understanding of molecular mechanisms for detoxification, metabolism, toxicity, and adaptation of fish. Previous studies with fish show modulation of expression of key genes in relation to stress response against WAF exposure, but there is still a lack of studies on responses of cytochrome P450 (CYP) genes and changes in biotransformation upon WAF exposure. In this study, we used the full spectrum of CYP genes of the marine medaka, Oryzias melastigma, to understand their potential mode of action on WAFs-triggered molecular mechanisms. We also analyzed further CYP-involved detoxification and endogenous steroidogenic metabolism after exposure to different concentrations of WAFs over different time courses in the marine medaka. Also, detoxification- and antioxidant-related enzymes' activities were analyzed with different concentrations of WAFs. As a result, the WAF exposure induced CYP-involved detoxification mechanism but reduced CYP-involved steroidogenic metabolism in the marine medaka. These data suggest that whole CYP profiling can be a way of understanding and uncovering the mode of action particularly with respect to emerging chemicals such as WAF exposure with the new finding that WAFs have dual functions on CYP-involved metabolisms.
Collapse
Affiliation(s)
- Jae-Sung Rhee
- Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, South Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Dash A, Simmers MB, Deering TG, Berry DJ, Feaver RE, Hastings NE, Pruett TL, LeCluyse EL, Blackman BR, Wamhoff BR. Hemodynamic flow improves rat hepatocyte morphology, function, and metabolic activity in vitro. Am J Physiol Cell Physiol 2013; 304:C1053-63. [PMID: 23485712 DOI: 10.1152/ajpcell.00331.2012] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In vitro primary hepatocyte systems typically elicit drug induction and toxicity responses at concentrations much higher than corresponding in vivo or clinical plasma C(max) levels, contributing to poor in vitro-in vivo correlations. This may be partly due to the absence of physiological parameters that maintain metabolic phenotype in vivo. We hypothesized that restoring hemodynamics and media transport would improve hepatocyte architecture and metabolic function in vitro compared with nonflow cultures. Rat hepatocytes were cultured for 2 wk either in nonflow collagen gel sandwiches with 48-h media changes or under controlled hemodynamics mimicking sinusoidal circulation within a perfused Transwell device. Phenotypic, functional, and metabolic parameters were assessed at multiple times. Hepatocytes in the devices exhibited polarized morphology, retention of differentiation markers [E-cadherin and hepatocyte nuclear factor-4α (HNF-4α)], the canalicular transporter [multidrug-resistant protein-2 (Mrp-2)], and significantly higher levels of liver function compared with nonflow cultures over 2 wk (albumin ~4-fold and urea ~5-fold). Gene expression of cytochrome P450 (CYP) enzymes was significantly higher (fold increase over nonflow: CYP1A1: 53.5 ± 10.3; CYP1A2: 64.0 ± 15.1; CYP2B1: 15.2 ± 2.9; CYP2B2: 2.7 ± 0.8; CYP3A2: 4.0 ± 1.4) and translated to significantly higher basal enzyme activity (device vs. nonflow: CYP1A: 6.26 ± 2.41 vs. 0.42 ± 0.015; CYP1B: 3.47 ± 1.66 vs. 0.4 ± 0.09; CYP3A: 11.65 ± 4.70 vs. 2.43 ± 0.56) while retaining inducibility by 3-methylcholanthrene and dexamethasone (fold increase over DMSO: CYP1A = 27.33 and CYP3A = 4.94). These responses were observed at concentrations closer to plasma levels documented in vivo in rats. The retention of in vivo-like hepatocyte phenotype and metabolic function coupled with drug response at more physiological concentrations emphasizes the importance of restoring in vivo physiological transport parameters in vitro.
Collapse
Affiliation(s)
- A Dash
- HemoShear, LLC, Charlottesville, VA 22902, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Kirby B, Kharasch ED, Thummel KT, Narang VS, Hoffer CJ, Unadkat JD. Simultaneous Measurement of In Vivo P-glycoprotein and Cytochrome P450 3A Activities. J Clin Pharmacol 2013; 46:1313-9. [PMID: 17050796 DOI: 10.1177/0091270006292625] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Digoxin and midazolam are routinely used as probe drugs to measure in vivo activity of P-glycoprotein (P-gp) and cytochrome P450 3A4/5 (CYP3A), respectively. We investigated whether digoxin and midazolam could be coadministered to simultaneously determine P-gp and CYP3A activity without a significant pharmacokinetic interaction. In a randomized crossover design, digoxin (0.5 mg oral) or midazolam (2.0 mg oral) was administered individually or in combination (digoxin 1 hour after midazolam) to 14 healthy volunteers. Blood and urine samples were collected for up to 48 hours. Pharmacokinetic parameters of digoxin, midazolam and 1'-OH midazolam were evaluated to determine the presence of an interaction. The geometric mean ratios of all measured pharmacokinetic parameters of digoxin and midazolam were not significantly affected by coadministration. Coadministration of digoxin and midazolam can be used to simultaneously phenotype P-gp and CYP3A activity without a significant pharmacokinetic interaction.
Collapse
Affiliation(s)
- Brian Kirby
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195-7610, USA
| | | | | | | | | | | |
Collapse
|
24
|
Areskog M, von Samson-Himmelstjerna G, Alvinerie M, Sutra JF, Höglund J. Dexamethasone treatment interferes with the pharmacokinetics of ivermectin in young cattle. Vet Parasitol 2012; 190:482-8. [PMID: 22959189 DOI: 10.1016/j.vetpar.2012.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 07/06/2012] [Accepted: 07/12/2012] [Indexed: 12/29/2022]
Abstract
An experiment was carried out to study the possible interaction between dexamethasone (DXM) treatment and the efficacy of ivermectin (IVM) treatment in young cattle. Two groups, each of seven calves, were experimentally inoculated with an equal mixture containing 15,000 third stage larvae of Cooperia oncophora and Ostertagia ostertagi each, and with no history of being resistant to any anthelmintics. However, in this study C. oncophora was unexpectedly classified as IVM-resistant according to the outcome from the faecal egg count reduction test (FECRT). Blood parameters and faecal egg counts (FEC) were monitored from 0 to 35 days post infection (d.p.i.). The calves in one group received intramuscular injections of short and long-term acting DXM at 22 and 24 d.p.i., respectively. The other group remained as a control. Three days post patency (24 d.p.i.) both groups were injected subcutaneously with IVM (Merial) at the recommended dose (0.2mg/kg). A significant difference (p<0.001) in FEC patterns was observed between groups. Although both groups still excreted eggs (100-200 eggs per gram faeces) 11 days post anthelmintic treatment, the control group had a significantly higher reduction between 23 and 35 d.p.i. (p=0.025). After 35 days, four animals per group were euthanized, and worms in the gastrointestinal tract were counted. No O. ostertagi were found in the abomasums, but low to high numbers (800-6200) of C. oncophora remained in the small intestines in both groups. Overall, these findings indicated that there was an interaction between the efficacy of IVM and DXM treatment. As significantly lower plasma levels of IVM were observed in the DXM group, we conclude that the impaired efficacy of ivermectin was most likely due to the altered pharmacokinetics.
Collapse
Affiliation(s)
- Marlene Areskog
- Department of Biomedical Sciences and Veterinary Public Health, Section for Parasitology, Swedish University of Agricultural Sciences, SE-751 89 Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
25
|
Kosaka K, Sakai N, Endo Y, Fukuhara Y, Tsuda-Tsukimoto M, Ohtsuka T, Kino I, Tanimoto T, Takeba N, Takahashi M, Kume T. Impact of intestinal glucuronidation on the pharmacokinetics of raloxifene. Drug Metab Dispos 2011; 39:1495-502. [PMID: 21646435 DOI: 10.1124/dmd.111.040030] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Raloxifene is extensively glucuronidated in humans, effectively reducing its oral bioavailability (2%). It was also reported to be glucuronidated in preclinical animals, but its effects on the oral bioavailability have not been fully elucidated. In the present study, raloxifene and its glucuronides in the portal and systemic blood were monitored in Gunn rats deficient in UDP-glucuronosyltransferase (UGT) 1A, Eisai hyperbilirubinemic rats (EHBRs), which hereditarily lack multidrug resistance-associated protein (MRP) 2, and wild-type rats after oral administration. The in vitro-in vivo correlation (IVIVC) of four UGT substrates (raloxifene, biochanin A, gemfibrozil, and mycophenolic acid) in rats was also evaluated. In Gunn rats, the product of fraction absorbed and intestinal availability and hepatic availability of raloxifene were 0.63 and 0.43, respectively; these values were twice those observed in wild-type Wistar rats, indicating that raloxifene was glucuronidated in both the liver and intestine. The ratio of glucuronides to unchanged drug in systemic blood was substantially higher in EHBRs (129-fold) than in the wild-type Sprague-Dawley rats (10-fold), suggesting the excretion of raloxifene glucuronides caused by MRP2. The IVIVC of the other UGT substrates in rats displayed a good relationship, but the oral clearance values of raloxifene and biochanin A, which were extensively glucuronidated by rat intestinal microsomes, were higher than the predicted clearances using rat liver microsomes, suggesting that intestinal metabolism may be a great contributor to the first-pass effect. Therefore, evaluation of intestinal and hepatic glucuronidation for new chemical entities is important to improve their pharmacokinetic profiles.
Collapse
Affiliation(s)
- Keigo Kosaka
- DMPK Research Laboratory, Mitsubishi Tanabe Corporation, 2-2-50, Kawagishi, Toda-shi, Saitama, 335-8505, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hofmeister CC, Yang X, Pichiorri F, Chen P, Rozewski DM, Johnson AJ, Lee S, Liu Z, Garr CL, Hade EM, Ji J, Schaaf LJ, Benson DM, Kraut EH, Hicks WJ, Chan KK, Chen CS, Farag SS, Grever MR, Byrd JC, Phelps MA. Phase I trial of lenalidomide and CCI-779 in patients with relapsed multiple myeloma: evidence for lenalidomide-CCI-779 interaction via P-glycoprotein. J Clin Oncol 2011; 29:3427-34. [PMID: 21825263 DOI: 10.1200/jco.2010.32.4962] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Multiple myeloma (MM) is an incurable plasma-cell neoplasm for which most treatments involve a therapeutic agent combined with dexamethasone. The preclinical combination of lenalidomide with the mTOR inhibitor CCI-779 has displayed synergy in vitro and represents a novel combination in MM. PATIENTS AND METHODS A phase I clinical trial was initiated for patients with relapsed myeloma with administration of oral lenalidomide on days 1 to 21 and CCI-779 intravenously once per week during a 28-day cycle. Pharmacokinetic data for both agents were obtained, and in vitro transport and uptake studies were conducted to evaluate potential drug-drug interactions. RESULTS Twenty-one patients were treated with 15 to 25 mg lenalidomide and 15 to 20 mg CCI-779. The maximum-tolerated dose (MTD) was determined to be 25 mg lenalidomide with 15 mg CCI-779. Pharmacokinetic analysis indicated increased doses of CCI-779 resulted in statistically significant changes in clearance, maximum concentrations, and areas under the concentration-time curves, with constant doses of lenalidomide. Similar and significant changes for CCI-779 pharmacokinetics were also observed with increased lenalidomide doses. Detailed mechanistic interrogation of this pharmacokinetic interaction demonstrated that lenalidomide was an ABCB1 (P-glycoprotein [P-gp]) substrate. CONCLUSION The MTD of this combination regimen was 25 mg lenalidomide with 15 mg CCI-779, with toxicities of fatigue, neutropenia, and electrolyte wasting. Pharmacokinetic and clinical interactions between lenalidomide and CCI-779 seemed to occur, with in vitro data indicating lenalidomide was an ABCB1 (P-gp) substrate. To our knowledge, this is the first report of a clinically significant P-gp-based drug-drug interaction with lenalidomide.
Collapse
|
27
|
Pałasz A, Wiaderkiewicz A, Wiaderkiewicz R, Czekaj P, Czajkowska B, Lebda-Wyborny T, Piwowarczyk A, Bryzek A. Age-related changes in the mRNA levels of CYP1A1, CYP2B1/2 and CYP3A1 isoforms in rat small intestine. GENES AND NUTRITION 2011; 7:197-207. [PMID: 21769602 DOI: 10.1007/s12263-011-0240-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 07/06/2011] [Indexed: 11/27/2022]
Abstract
It has been established beyond doubt that, as well as the liver, the small intestine is an important site of first-pass metabolism of numerous drugs, food components and toxic xenobiotics. However, there is not much information available about age-dependent changes of intestinal biotransformation pathways. In the present paper, we evaluated the relationships between intestinal cytochrome P450 complex activity and the age of animals. The study was carried out on male Sprague-Dawley rats (n = 5) from 5 age series: 0.5-, 2-, 4-, 20-, and 28 months old. Animals at every age series were divided into 4 groups: control and three groups of rats treated with the CYP450 specific inducers: phenobarbital, β-naphtoflavone and dexamethasone, respectively. RNA was isolated from intestinal mucosa, and then standard RT-PCR was used for the analysis of CYP1A1, CYP2B1/2 and CYP3A1 mRNA expression. Additionally, the activities of NADPH-cytochrome P450 and NADH-cytochrome b(5) reductases in the microsomal fraction were biochemically estimated. The constitutive intestinal CYP1A1 mRNA expression changes during maturation and aging. Inducibility of CYP1A1 gene was evident in intestinal mucosa at 2-, 4- and 20-month-old rats. A similar pattern of changes was observed for CYP2B1/2 isoforms. CYP3A1 mRNA expression was not detected in small intestine of 2-week-old rats. In matured rats, constitutive intestinal CYP3A1 expression was low, although after induction, significant increases in CYP3A1 mRNA amount were noted in aged individuals. Intestinal activity of both analyzed reductases was lowest in immature rats and highest in 28-month-old animals. In conclusion, the activity of cytochrome P450 complex in rat small intestine was not decreased by the aging processes, so the high rate of oxidative metabolic reactions in intestinal mucosa can be maintained till the advanced life stage.
Collapse
Affiliation(s)
- Artur Pałasz
- Department of Histology, Medical University of Silesia, ul. Medyków 18, 40-752, Katowice, Poland,
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abuznait AH, Patrick SG, Kaddoumi A. Exposure of LS-180 cells to drugs of diverse physicochemical and therapeutic properties up-regulates P-glycoprotein expression and activity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2011; 14:236-48. [PMID: 21733412 PMCID: PMC3172709 DOI: 10.18433/j36016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Drug transporters are increasingly recognized as important determinants of variability in drug disposition and therapeutic response, both in pre-clinical and clinical stages of drug development process. The role P-glycoprotein (P-gp) plays in drug interactions via its inhibition is well established. However, much less knowledge is available about drugs effect on P-gp up-regulation. The objective of this work was to in vitro investigate and rank commonly used drugs according to their potencies to up-regulate P-gp activity utilizing the same experimental conditions. METHODS The in vitro potencies of several drugs of diverse physicochemical and therapeutic properties including rifampicin, dexamethasone, caffeine, verapamil, pentylenetetrazole, hyperforin, and β-estradiol over broad concentration range to up-regulate P-gp expression and activity were examined. For dose-response studies, LS-180 cells were treated with different concentrations of the selected drugs followed by P-gp protein and gene expressions analyses. P-gp functionality was determined by uptake studies with rhodamine 123 as a P-gp substrate, followed by Emax/EC50 evaluation. RESULTS The results demonstrated a dose-dependent increase in P-gp expression and activity following treatments. At 50 uM concentration (hyperforin, 0.1 uM), examined drugs increased P-gp protein and gene expressions by up to 5.5 and 6.2-fold, respectively, while enhanced P-gp activity by 1.8-4-fold. The rank order of these drugs potencies to up-regulate P-gp activity was as following: hyperforin >>> dexamethasone ~ beta-estradiol > caffeine > rifampicin ~ pentylenetetrazole > verapamil. CONCLUSIONS These drugs have the potential to be involved in drug interactions when administered with other drugs that are P-gp substrates. Further studies are needed to in vivo evaluate these drugs and verify the consequences of such induction on P-gp activity for in vitro-in vivo correlation purposes.
Collapse
Affiliation(s)
- Alaa H Abuznait
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, USA
| | | | | |
Collapse
|
29
|
Shen Y, Zhu RH, Li HD, Liu YW, Xu P. Validated LC–MS (ESI) assay for the simultaneous determination of amitriptyline and its metabolite nortriptyline in rat plasma: Application to a pharmacokinetic comparison. J Pharm Biomed Anal 2010; 53:735-9. [DOI: 10.1016/j.jpba.2010.04.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 04/13/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
|
30
|
Kirschbaum KM, Uhr M, Holthoewer D, Namendorf C, Pietrzik C, Hiemke C, Schmitt U. Pharmacokinetics of acute and sub-chronic aripiprazole in P-glycoprotein deficient mice. Neuropharmacology 2010; 59:474-9. [DOI: 10.1016/j.neuropharm.2010.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 06/21/2010] [Accepted: 06/24/2010] [Indexed: 12/20/2022]
|
31
|
Transactivation of genes encoding for phase II enzymes and phase III transporters by phytochemical antioxidants. Molecules 2010; 15:6332-48. [PMID: 20877225 PMCID: PMC6257698 DOI: 10.3390/molecules15096332] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Revised: 08/31/2010] [Accepted: 09/03/2010] [Indexed: 01/27/2023] Open
Abstract
The induction of phase II enzymes and phase III transporters contributes to the metabolism, detoxification of xenobiotics, antioxidant capacity, redox homeostasis and cell viability. Transactivation of the genes that encode for phase II enzymes and phase III transporters is coordinatively regulated by activating transcription factors in response to external stimuli. Comprehensive studies indicate that antioxidant phytochemicals promote the induction of phase II enzymes and/or phase III transporters through various signaling pathways, including phosphoinositide 3-kinase, protein kinase C, and mitogen-activated protein kinases. This paper focuses on the molecular mechanisms and signaling pathways responsible for the transactivation of genes encoding for these proteins, as orchestrated by a series of transcription factors and related signaling components.
Collapse
|
32
|
Vautier S, Fernandez C. ABCB1: the role in Parkinson's disease and pharmacokinetics of antiparkinsonian drugs. Expert Opin Drug Metab Toxicol 2010; 5:1349-58. [PMID: 19663741 DOI: 10.1517/17425250903193079] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
ABCB1/P-glycoprotein (P-gp) is an ATP-dependant transmembrane efflux protein widely expressed in human organs and plays a protective role against endogenous and exogenous substances. It is involved in drug pharmacokinetics affecting drug absorption, disposition and elimination. At the BBB level, due to its luminal localisation, ABCB1 limits drug transport and is important in central detoxification. Inter-individual variability has been described in ABCB1 expression and functionality. Recent work suggests that variability may play a role in the pathogenesis of neurological diseases. Furthermore, ABCB1 expression and/or functionality may modify drug efficacy or increase central adverse events. This paper reviews ABCB1 implication in the pathophysiology of Parkinson's disease and its role in the cerebral distribution of drugs.
Collapse
Affiliation(s)
- Sarah Vautier
- University Paris-Sud XI, Department of Clinical Pharmacy, Chatenay-Malabry, France.
| | | |
Collapse
|
33
|
The complexities of antiretroviral drug-drug interactions: role of ABC and SLC transporters. Trends Pharmacol Sci 2009; 31:22-35. [PMID: 20004485 DOI: 10.1016/j.tips.2009.10.001] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 09/30/2009] [Accepted: 10/01/2009] [Indexed: 12/15/2022]
Abstract
Treatment of human immunodeficiency virus (HIV) infection involves a combination of several antiviral agents belonging to different pharmacological classes. This combination is referred to as highly active antiretroviral therapy (HAART). This treatment has proved to be very effective in suppressing HIV replication, but antiretroviral drugs have complex pharmacokinetic properties involving extensive drug metabolism and transport by membrane-associated drug carriers. Combination drug therapy often introduces complex drug-drug interactions that can result in toxic or sub-therapeutic drug concentrations, compromising treatment. This review focuses on the role of ATP-binding cassette (ABC) membrane-associated efflux transporters and solute carrier (SLC) uptake transporters in antiretroviral drug disposition, and identifies clinically important antiretroviral drug-drug interactions associated with changes in drug transport.
Collapse
|
34
|
Zastre JA, Chan GNY, Ronaldson PT, Ramaswamy M, Couraud PO, Romero IA, Weksler B, Bendayan M, Bendayan R. Up-regulation of P-glycoprotein by HIV protease inhibitors in a human brain microvessel endothelial cell line. J Neurosci Res 2009; 87:1023-36. [PMID: 18855943 DOI: 10.1002/jnr.21898] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A major concern regarding the chronic administration of antiretroviral drugs is the potential for induction of drug efflux transporter expression (i.e., P-glycoprotein, P-gp) at tissue sites that can significantly affect drug distribution and treatment efficacy. Previous data have shown that the inductive effect of human immunodeficiency virus protease inhibitors (PIs) is mediated through the human orphan nuclear receptor, steroid xenobiotic receptor (SXR or hPXR). The objectives of this study were to investigate transport and inductive properties on efflux drug transporters of two PIs, atazanavir and ritonavir, at the blood-brain barrier by using a human brain microvessel endothelial cell line, hCMEC/D3. Transport properties of PIs by the drug efflux transporters P-gp and multidrug resistance protein 1 (MRP1) were assessed by measuring the cellular uptake of (3)H-atazanavir or (3)H-ritonavir in P-gp and MRP1 overexpressing cells as well as hCMEC/D3. Whereas the P-gp inhibitor, PSC833, increased atazanavir and ritonavir accumulation in hCMEC/D3 cells by 2-fold, the MRP inhibitor MK571 had no effect. P-gp, MRP1, and hPXR expression and localization were examined by Western blot analysis and immunogold cytochemistry at the electron microscope level. Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. In hCMEC/D3 cells, cellular accumulation of these PIs appears to be primarily limited by P-gp efflux activity. Long-term exposure of atazanavir or ritonavir to brain microvessel endothelium may result in further limitations in brain drug permeability as a result of the up-regulation of P-gp expression and function.
Collapse
Affiliation(s)
- Jason A Zastre
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kharasch ED, Bedynek PS, Walker A, Whittington D, Hoffer C. Mechanism of ritonavir changes in methadone pharmacokinetics and pharmacodynamics: II. Ritonavir effects on CYP3A and P-glycoprotein activities. Clin Pharmacol Ther 2009; 84:506-12. [PMID: 19238656 DOI: 10.1038/clpt.2008.102] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. We determined short-term (2-day) and steady-state (2-week) ritonavir effects on intestinal and hepatic CYP3A4/5 (probed with intravenous (IV) and oral alfentanil (ALF) and with miosis) and P-glycoprotein (P-gp) (fexofenadine), and on methadone pharmacokinetics and pharmacodynamics in healthy volunteers. Acute ritonavir increased the area under the concentration-time curve (AUC)(0-infinity)/dose ratio (ritonavir/control) for oral ALF 25-fold. Steady-state ritonavir increased the AUC(0-Infinity)/dose ratio for IV and oral ALF 4- and 10-fold, respectively; reduced hepatic extraction (from 0.26 to 0.07) and intestinal extraction (from 0.51 to 0); and increased bioavailability (from 37 to 95%). Acute ritonavir inhibits first-pass CYP3A > 96%. Chronic ritonavir inhibits hepatic CYP3A (> 70%) and first-pass CYP3A (> 90%). Acute and steady-state ritonavir increased the fexofenadine AUC(0-infinity) 2.8- and 1.4-fold, respectively, suggesting P-gp inhibition. Steady-state compared with acute ritonavir caused mild apparent induction of P-gp and hepatic CYP3A, but net inhibition still predominated. Ritonavir inhibited both intestinal and hepatic CYP3A and drug transport. ALF miosis noninvasively determined CYP3A inhibition by ritonavir.
Collapse
Affiliation(s)
- E D Kharasch
- Division of Clinical and Translational Research, Department of Anesthesiology, Washington University, St. Louis, Missouri, USA.
| | | | | | | | | |
Collapse
|
36
|
Kharasch ED, Bedynek PS, Park S, Whittington D, Walker A, Hoffer C. Mechanism of ritonavir changes in methadone pharmacokinetics and pharmacodynamics: I. Evidence against CYP3A mediation of methadone clearance. Clin Pharmacol Ther 2009; 84:497-505. [PMID: 19238655 DOI: 10.1038/clpt.2008.104] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ritonavir diminishes methadone plasma concentrations, an effect attributed to CYP3A induction, but the actual mechanisms are unknown. We determined ritonavir effects on stereoselective methadone pharmacokinetics and clinical effects (pupillary miosis) in healthy human immunodeficiency virus-negative volunteers. Subjects received intravenous plus oral (deuterium-labeled) racemic methadone after no ritonavir, short-term (3-day) ritonavir, and steady-state ritonavir. Acute and steady-state ritonavir, respectively, caused 1.5- and 2-fold induction of systemic and apparent oral R- and S-methadone clearances. Ritonavir increased renal clearance 40-50%, and stereoselectively (S > R) increased hepatic methadone N-demethylation 50-80%, extraction twofold, and clearance twofold. Bioavailability was unchanged despite significant inhibition of intestinal P-glycoprotein. Intestinal and hepatic CYP3A was inhibited > 70%. Ritonavir shifted methadone plasma concentration-miosis curves leftward and upward. Rapid ritonavir induction of methadone clearance results from increased renal clearance and induced hepatic metabolism. Induction of methadone metabolism occurred despite profound CYP3A inhibition, suggesting no role for CYP3A in clinical methadone metabolism and clearance. Ritonavir may alter methadone pharmacodynamics.
Collapse
Affiliation(s)
- E D Kharasch
- Division of Clinical and Translational Research, Department of Anesthesiology, Washington University, St. Louis, Missouri, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Milane A, Vautier S, Chacun H, Meininger V, Bensimon G, Farinotti R, Fernandez C. Interactions between riluzole and ABCG2/BCRP transporter. Neurosci Lett 2009; 452:12-6. [PMID: 19146924 DOI: 10.1016/j.neulet.2008.12.061] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 12/19/2008] [Accepted: 12/31/2008] [Indexed: 01/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative fatal disease. Drugs used in this disease need to cross the blood-brain barrier (BBB). Only riluzole is approved for ALS treatment. We have investigated riluzole as a breast cancer resistance protein (BCRP) substrate by studying its brain transport in CF1 mdr1a (-/-) mice and its intracellular uptake on BeWo cells (human placental choriocarcinoma cell line). We have also investigated the effect of riluzole on BCRP expression level and on its activity using the prazocin as a test probe for brain transport and intracellular uptake. Assays on mdr1a (-/-) mice and BeWo cells showed a higher uptake of riluzole when pretreated with a BCRP inhibitor. After repeated doses of riluzole, BCRP activity was increased in CF1 mdr1a (-/-) mice, riluzole uptake was decrease and both BCRP expression and activity were increased in BeWo cells. In conclusion, we report in this study that riluzole is transported by BCRP at the BBB level and can enhance its function. These results taken with our previous studies on riluzole and P-glycoprotein show that drug-drug interactions between riluzole and efflux transporters substrates may occur at the BBB level and should be taken into account in future clinical trial design in ALS.
Collapse
Affiliation(s)
- Aline Milane
- Université Paris-Sud XI, Laboratoire de Barrières et Passage des Médicaments, EA 2706, Faculty of Pharmacy, Chatenay-Malabry, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Ritonavir Greatly Impairs CYP3A Activity in HIV Infection With Chronic Viral Hepatitis. J Acquir Immune Defic Syndr 2008; 49:358-68. [DOI: 10.1097/qai.0b013e31818c7efe] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Micuda S, Fuksa L, Brcakova E, Osterreicher J, Cermanova J, Cibicek N, Mokry J, Staud F, Martinkova J. Zonation of multidrug resistance-associated protein 2 in rat liver after induction with dexamethasone. J Gastroenterol Hepatol 2008; 23:e225-30. [PMID: 17683490 DOI: 10.1111/j.1440-1746.2007.05066.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIM The present study was aimed to evaluate the hepatic zonation of multidrug resistance-associated protein 2 (mrp2), an important drug transporter, and its potential changes during the induction of its expression by known inducer, dexamethasone (DEX). METHODS The hepatic expression of mrp2 was studied by immunohistochemistry with consequent quantification by measurement of integral optical densities of mrp2 staining in the periportal and perivenous areas of the liver acinus in control and DEX-pretreated rats (1 mg/kg daily per os for 4 days). Overall changes in mrp2 expression and function produced by DEX were monitored using Western blotting and an in vivo clearance study of endogenous-conjugated bilirubin, a mrp2 substrate. RESULTS In the control animals, a quantitative image analysis revealed the primary periportal localization of mrp2 within the liver acinus with the expression of mrp2 being 16.7-fold of that in the perivenous area. After DEX pretreatment, the expression of mrp2 increased, especially in the perivenous hepatocytes. The overall expression of mrp2 increased 3.2-fold in comparison with the control group. This observation was confirmed by Western blotting, which showed a 1.3-fold increase in the mrp2 protein after DEX pretreatment. The functional consequences of the induced mrp2 protein in the livers of the DEX-pretreated rats were demonstrated by the increased biliary excretion of conjugated bilirubin. CONCLUSION In conclusion, these results indicate the zonation of mrp2 protein expression primarily to periportal hepatocytes. The induction by DEX produced spatially disproportional changes with an increase in the mrp2 protein being most prominent in the perivenous hepatocytes.
Collapse
Affiliation(s)
- Stanislav Micuda
- Department of Pharmacology, Charles University in Prague, Faculty of Medicine in Hradec Kralove, Simkova, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fukushima K, Haraya K, Terasaka S, Ito Y, Sugioka N, Takada K. Long-term pharmacokinetic efficacy and safety of low-dose ritonavir as a booster and atazanavir pharmaceutical formulation based on solid dispersion system in rats. Biol Pharm Bull 2008; 31:1209-14. [PMID: 18520056 DOI: 10.1248/bpb.31.1209] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atazanavir (ATV) is clinically coadministered with low-dose ritonavir (RTV), which boosts the oral bioavailability (BA) of ATV by inhibiting cytochrome P450 (CYP) 3A, and P-glycoprotein (Pgp) via the same metabolic pathway; however, it is well known that in the chronic phase, the inhibition effect of RTV on Pgp and CYP3A becomes an induction effect. In this study, we investigated the long-term efficacy and safety of RTV-boosted ATV in rats with a clinical relevant dosage of ATV and RTV, 7 mg/kg and 2 mg/kg, respectively, and drew a direct comparison with RTV-boosted ATV and the previously reported ATV pharmaceutical formulation based on a solid dispersion system (ATV-SLS SD+G). Rats received RTV-boosted ATV or ATV-SLS SD+G for 14 d in the pharmacokinetic study. In addition, after 14-d repeated administration of each formulation, cyclosporine A (CyA) was administered to rats and Western blot analysis of Pgp and CYP3A was performed to investigate the impact on pharmacokinetic interaction of each ATV formulation. After repeated administration of both formulations, there was no significant difference between ATV pharmacokinetic parameters on day 1 and 14; therefore, it was considered that the long-term efficacy of both ATV formulations was maintained. However, after treatment with RTV-boosted ATV, the Cmax and AUC0-infinity of the following CyA significantly decreased to 49% and 47% in comparison to the control, respectively, and the Pgp expression in the small intestine by Western blot analysis was approximately 2-fold higher than the control, whereas after treatment with ATV pharmaceutical formulation, neither significant alteration of CyA nor notable change in the expression of intestinal Pgp and hepatic CYP3A was observed. Therefore, it was considered that the BA of CyA after treatment with RTV-boosted ATV would decrease by the induction effect of RTV in chronic phase as described above. The results of this study revealed that the chronic use of low-dose RTV as a booster has great potential to compromise drug-drug interactions; therefore, it is recommended that the BA of protease inhibitors be improved by a pharmaceutical approach without pharmacokinetic interaction by RTV.
Collapse
Affiliation(s)
- Keizo Fukushima
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto, Japan.
| | | | | | | | | | | |
Collapse
|
41
|
Narang VS, Fraga C, Kumar N, Shen J, Throm S, Stewart CF, Waters CM. Dexamethasone increases expression and activity of multidrug resistance transporters at the rat blood-brain barrier. Am J Physiol Cell Physiol 2008; 295:C440-50. [PMID: 18524938 DOI: 10.1152/ajpcell.00491.2007] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Brain edema is an important factor leading to morbidity and mortality associated with primary brain tumors. Dexamethasone, a synthetic glucocorticoid, is routinely prescribed with antineoplastic agents to alleviate pain associated with chemotherapy and reduce intracranial pressure. We investigated whether dexamethasone treatment increased the expression and activity of multidrug resistance (MDR) transporters at the blood-brain barrier. Treatment of primary rat brain microvascular endothelial cells with submicromolar concentrations of dexamethasone induced significantly higher levels of drug efflux transporters such as breast cancer resistance protein (abcg2), P-glycoprotein (P-gp; abcb1a/abcb1b), and MDR protein 2 (Mrp2; abcc2) as indicted by protein and mRNA levels as well as by functional activity. The effect of dexamethasone on transporter function was significant within 6 h of treatment, was dose dependent, and was reversible. Dexamethasone-induced upregulation of Bcrp and P-gp expression and function was partially abrogated by the glucocorticoid receptor (GR) antagonist RU486. In contrast, RU486 had no effect on the dexamethasone-induced upregulation of Mrp2, suggesting a GR-independent regulation of Mrp2, and a GR-dependent regulation of P-gp and Bcrp. In addition to the dexamethasone-induced upregulation of MDR transporters, we measured a dose-dependent and reversible increase in the expression of the nuclear transcription factor pregnane xenobiotic receptor (PXR). Administering dexamethasone to rats caused increased expression of PXR in brain microvessels within 24 h. These results suggest that adjuvant therapy with corticosteroids such as dexamethasone in the treatment of brain tumors may increase the expression of MDR transporters at the blood-brain barrier through pathways involving GR and PXR.
Collapse
Affiliation(s)
- Vishal S Narang
- Dept. of Physiology, Univ. of Tennessee Health Science Center, 894 Union Ave., Nash 426, Memphis, TN 38163, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Gupta A, Mugundu GM, Desai PB, Thummel KE, Unadkat JD. Intestinal human colon adenocarcinoma cell line LS180 is an excellent model to study pregnane X receptor, but not constitutive androstane receptor, mediated CYP3A4 and multidrug resistance transporter 1 induction: studies with anti-human immunodeficiency virus protease inhibitors. Drug Metab Dispos 2008; 36:1172-80. [PMID: 18332086 DOI: 10.1124/dmd.107.018689] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Lack of an established cell line model to study induction of cytochromes P450 (P450s) and drug transporters poses a challenge in predicting in vivo drug-drug interactions. Although not well characterized, LS180 cells could be an excellent cell line to study induction of P450s and transporters because they express pregnane X receptor (PXR). Therefore, as part of a larger study of in vitro to in vivo prediction of inductive drug interactions, we determined induction of various P450s and drug transporters by the anti-human deficiency virus protease inhibitors (PIs) and the prototypic inducer, rifampin, in LS180 cells. Among these proteins, the various PIs significantly induced (n = 3-5) only CYP3A4 and multidrug resistance transporter 1 (MDR1) transcripts (2- to 50-fold). CYP3A4 activity (1'-hydroxymidazolam formation) was increased (2-fold) by rifampin (10 microM) but was reduced by the PIs (1.5- to 7-fold). Surprisingly, constitutive androstane receptor 1 (CAR1) was not found to be expressed in these cells. Additionally, using a reporter assay, we found that PIs did not activate CAR3 (the natural splice variant of CAR1) but significantly activated PXR (2- to 24-fold), which correlated well with induction of CYP3A4 and MDR1 transcripts (approximately r = 0.9). Furthermore, in a PXR-knockdown stable LS180 cell line, induction of CYP3A4 and MDR1 mRNA after treatment with PIs and rifampin was significantly reduced (1.4- to 5-fold) compared with that in PXR nonsilenced cells. Based on these data, we conclude that LS180 cells could be used as a readily available, high-throughput cell line to screen for PXR-mediated induction of CYP3A4 and MDR1 transcripts. These data also indicate that the majority of the PIs are likely to produce intestinal drug-drug interactions by inactivating or inhibiting CYP3A enzymes even though they induce CYP3A4 and MDR1 transcripts via PXR.
Collapse
Affiliation(s)
- Anshul Gupta
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | |
Collapse
|
43
|
Perloff MD, von Moltke LL, Fahey JM, Greenblatt DJ. Induction of P-glycoprotein expression and activity by ritonavir in bovine brain microvessel endothelial cells. J Pharm Pharmacol 2007; 59:947-53. [PMID: 17637189 DOI: 10.1211/jpp.59.7.0006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Extended treatment with human immunodeficiency virus (HIV) protease inhibitors (HPIs) is standard in HIV/AIDS therapy. While these drugs have helped decrease the overall incidence of AIDS defining illnesses, the relative prevalence of HIV/AIDS dementia has increased. HPIs may cause induction of blood-brain barrier (BBB) drug transporters (P-glycoprotein; P-gp) and thereby limit entry of HPIs into brain tissue, increasing the probability that the brain could become an HIV sanctuary site. Using bovine brain microvessel endothelial cells (BMEC) as an in-vitro model of the BBB, the potential for the HIV protease inhibitor ritonavir to cause induction of P-gp activity and expression was examined. BMEC were isolated from fresh cow brain by enzymatic digest and density centrifugation. Primary culture BMEC were co-incubated with ritonavir or vehicle control for 120 h. Quantitative drug accumulation of rhodamine 123 (Rh123) and fluorescence microscopy were used as measures of P-gp activity. P-gp expression was assessed using quantitative Western blotting. Ritonavir decreased Rh123 cell accumulation and increased P-gp immunoreactive protein in a concentration-dependent manner. Fluorescent microscopy mirrored Rh123 quantitative studies. In BMEC pretreated with 30 microM ritonavir, Rh123 accumulation was decreased 40% and immunoreactive P-gp protein increased 2-fold. Collectively, a strong correlation between decreased Rh123 BMEC accumulation and increased P-gp immunoreactive protein was observed (Spearman r2 = 0.77, P < 0.0001). Thus extended exposure of BMEC to ritonavir caused a concentration-dependent increase in P-gp activity and expression. Similar findings may occur at the clinical level with prolonged HIV protease inhibitor use, giving insight into the central nervous system as an HIV sanctuary site and eventual development of HIV dementia.
Collapse
Affiliation(s)
- Michael D Perloff
- Department of Pharmacology and Experimental Therapeutics, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | |
Collapse
|
44
|
Lum PY, He YD, Slatter JG, Waring JF, Zelinsky N, Cavet G, Dai X, Fong O, Gum R, Jin L, Adamson GE, Roberts CJ, Olsen DB, Hazuda DJ, Ulrich RG. Gene expression profiling of rat liver reveals a mechanistic basis for ritonavir-induced hyperlipidemia. Genomics 2007; 90:464-73. [PMID: 17719200 DOI: 10.1016/j.ygeno.2007.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 06/16/2007] [Accepted: 06/19/2007] [Indexed: 11/26/2022]
Abstract
The molecular mechanisms of action of a HIV protease inhibitor, ritonavir, on hepatic function were explored on a genomic scale using microarrays comprising genes expressed in the liver of Sprague-Dawley rats (Rattus norvegicus). Analyses of hepatic transcriptional fingerprints led to the identification of several key cellular pathways affected by ritonavir treatment. These effects were compared to a compendium of gene expression responses for 52 unrelated compounds and to other protease inhibitors, including atazanavir and two experimental compounds. We identified genes involved in cholesterol and fatty acid biosynthesis, as well as genes involved in fatty acid and cholesterol breakdown, whose expressions were regulated in opposite manners by ritonavir and bezafibrate, a hypolipidemic agonist of the peroxisome proliferator-activated receptor alpha. Ritonavir also upregulated multiple proteasomal subunit transcripts as well as genes involved in ubiquitination, consistent with its known inhibitory effect on proteasomal activity. We also tested three other protease inhibitors in addition to ritonavir. Atazanavir did not impact ubiquitin or proteasomal gene expression, although the two other experimental protease inhibitors impacted both proteasomal gene expression and sterol regulatory element-binding protein-activated genes, similar to ritonavir. Identification of key metabolic pathways that are affected by ritonavir and other protease inhibitors will enable us to understand better the downstream effects of protease inhibitors, thus leading to better drug design and an effective method to mitigate the side effects of this important class of HIV therapeutics.
Collapse
Affiliation(s)
- Pek Yee Lum
- Rosetta Inpharmatics LLC, 401 Terry Avenue North, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wang X, Chai H, Yao Q, Chen C. Molecular mechanisms of HIV protease inhibitor-induced endothelial dysfunction. J Acquir Immune Defic Syndr 2007; 44:493-9. [PMID: 17245228 DOI: 10.1097/qai.0b013e3180322542] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Highly active antiretroviral therapy incorporating protease inhibitors (PIs) is successful in controlling HIV infection and has dramatically improved the prognosis of HIV-infected patients. The therapeutic benefit of long-term use of HIV PIs is compromised by an increased risk of cardiovascular disease, however, including metabolic syndrome and endothelial dysfunction. Although clinical evidence strongly suggests an association of the use of HIV PIs with endothelial dysfunction, the underlying molecular mechanisms have not been fully elucidated yet. In this review, we describe recent advances in the molecular mechanisms of PI-induced endothelial dysfunction. The available evidence demonstrates that certain HIV PIs could induce endothelial dysfunction, including a decrease of endothelium-dependent vasorelaxation, inhibition of the nitric oxide synthase system, increase of oxidative stress, and activation of mitogen-activated protein kinases. HIV infection itself may also induce endothelial dysfunction and injury. These new discoveries provide a better understanding of the molecular mechanisms of the interaction between HIV PIs and vascular cells and may suggest potential approaches to control HIV PI-associated cardiovascular complications.
Collapse
Affiliation(s)
- Xinwen Wang
- Molecular Surgeon Research Center, Division of Vascular Surgery and Endovascular Therapy, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
46
|
Abstract
Neurodegenerative and infectious disorders including Alzheimer's and Parkinson's diseases, amyotrophic lateral sclerosis, and stroke are rapidly increasing as population's age. Alzheimer's disease alone currently affects 4.5 million Americans, and more than $100 billion is spent per year on medical and institutional care for affected people. Such numbers will double in the ensuing decades. Currently disease diagnosis for all disorders is made, in large measure, on clinical grounds as laboratory and neuroimaging tests confirm what is seen by more routine examination. Achieving early diagnosis would enable improved disease outcomes. Drugs, vaccines or regenerative proteins present "real" possibilities for positively affecting disease outcomes, but are limited in that their entry into the brain is commonly restricted across the blood-brain barrier. This review highlights how these obstacles can be overcome by polymer science and nanotechnology. Such approaches may improve diagnostic and therapeutic outcomes. New developments in polymer science coupled with cell-based delivery strategies support the notion that diseases that now have limited therapeutic options can show improved outcomes by advances in nanomedicine.
Collapse
Affiliation(s)
- A.V. Kabanov
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - H.E. Gendelman
- Center for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
47
|
Zhang H, Cui D, Wang B, Han YH, Balimane P, Yang Z, Sinz M, Rodrigues AD. Pharmacokinetic Drug Interactions Involving 17??-Ethinylestradiol. Clin Pharmacokinet 2007; 46:133-57. [PMID: 17253885 DOI: 10.2165/00003088-200746020-00003] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
17alpha-Ethinylestradiol (EE) is widely used as the estrogenic component of oral contraceptives (OC). In vitro and in vivo metabolism studies indicate that EE is extensively metabolised, primarily via intestinal sulfation and hepatic oxidation, glucuronidation and sulfation. Cytochrome P450 (CYP)3A4-mediated EE 2-hydroxylation is the major pathway of oxidative metabolism of EE. For some time it has been known that inducers of drug-metabolising enzymes (such as the CYP3A4 inducer rifampicin [rifampin]) can lead to breakthrough bleeding and contraceptive failure. Conversely, inhibitors of drug-metabolising enzymes can give rise to elevated EE plasma concentrations and increased risks of vascular disease and hypertension. In vitro studies have also shown that EE inhibits a number of human CYP enzymes, such as CYP2C19, CYP3A4 and CYP2B6. Consequently, there are numerous reports in the literature describing EE-containing OC formulations as perpetrators of pharmacokinetic drug interactions. Because EE may participate in multiple pharmacokinetic drug interactions as either a victim or perpetrator, pharmaceutical companies routinely conduct clinical drug interaction studies with EE-containing OCs when evaluating new chemical entities in development. It is therefore critical to understand the mechanisms underlying these drug interactions. Such an understanding can enable the interpretation of clinical data and lead to a greater appreciation of the profile of the drug by physicians, clinicians and regulators. This article summarises what is known of the drug-metabolising enzymes and transporters governing the metabolism, disposition and excretion of EE. An effort is made to relate this information to known clinical drug-drug interactions. The inhibition and induction of drug-metabolising enzymes by EE is also reviewed.
Collapse
Affiliation(s)
- Hongjian Zhang
- Metabolism and Pharmacokinetics, Bristol-Myers Squibb Pharmaceutical Research Institute, Princeton, New Jersey 08543, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Maier A, Zimmermann C, Beglinger C, Drewe J, Gutmann H. Effects of budesonide on P-glycoprotein expression in intestinal cell lines. Br J Pharmacol 2006; 150:361-8. [PMID: 17179942 PMCID: PMC2013900 DOI: 10.1038/sj.bjp.0706992] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND AND PURPOSE P-glycoprotein (P-gp) is an important efflux transporter that supports the barrier function of the gut against invading antigens and against administered drugs. Since glucocorticoids, such as budesonide, are frequently used during inflammatory bowel disease we investigated how budesonide influences P-gp expression in different intestinal cell lines. EXPERIMENTAL APPROACH LS180 and Caco-2 cells were incubated with budesonide and changes in P-gp expression were determined on mRNA, protein and functional level. The mRNA expression levels of glucocorticoid receptor (GR) and pregnane X receptor (PXR) were determined in these cell lines. PXR receptor was transiently transfected into Caco-2 cells. KEY RESULTS Budesonide showed an induction of P-gp in LS180 cells and a down-regulation in Caco-2 cells. Expression levels of nuclear receptors revealed high expression of PXR only in LS180 cells and exclusive expression of GR in Caco-2 cells. Mifepristone, an anti-glucocorticoid, could not reverse the down-regulation of P-gp by budesonide in Caco-2 cells. In PXR-transfected Caco-2 cells the budesonide-mediated down-regulation of P-gp was abolished. Furthermore the expression of cytochrome P450 3A4 (CYP3A4), another PXR target gene, was induced in PXR-transfected Caco-2 cells after budesonide treatment. CONCLUSIONS AND IMPLICATIONS Budesonide has the potential to influence MDR1 expression in vitro. In LS180 cells, the induction of MDR1 by budesonide probably is mediated via PXR. The mechanism of the down-regulation in Caco-2 cells still remains unclear, but GR does not seem to be involved. Further studies are required to evaluate how budesonide alters P-gp expression in vivo.
Collapse
Affiliation(s)
- A Maier
- Department of Clinical Pharmacology and Toxicology, University Hospital Basel Basel, Switzerland
| | - C Zimmermann
- Department of Clinical Pharmacology and Toxicology, University Hospital Basel Basel, Switzerland
| | - C Beglinger
- Department of Internal Medicine, Division of Gastroenterology, University Hospital of Basel Basel, Switzerland
| | - J Drewe
- Department of Clinical Pharmacology and Toxicology, University Hospital Basel Basel, Switzerland
- Author for correspondence:
| | - H Gutmann
- Department of Clinical Pharmacology and Toxicology, University Hospital Basel Basel, Switzerland
| |
Collapse
|
49
|
van Heeswijk RPG, Bourbeau M, Campbell P, Seguin I, Chauhan BM, Foster BC, Cameron DW. Time-dependent interaction between lopinavir/ritonavir and fexofenadine. J Clin Pharmacol 2006; 46:758-67. [PMID: 16809801 DOI: 10.1177/0091270006288733] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This study investigated the effect of single-dose and steady-state lopinavir/ritonavir on the exposure to fexofenadine, as a measure of P-glycoprotein activity. Sixteen volunteers (8 women) received single-dose oral fexofenadine 120 mg alone, in combination with single-dose ritonavir 100 mg or lopinavir/ritonavir 400/100 mg (randomized 1:1, stratified by sex), and in combination with steady-state lopinavir/ritonavir 400/100 mg twice daily. Single-dose ritonavir and lopinavir/ritonavir increased the area under the fexofenadine plasma concentration-time curve from 0 to infinity (AUC(infinity)) by 2.2- and 4.0-fold, respectively (P < .02). Steady-state lopinavir/ritonavir increased the fexofenadine AUC(infinity) by 2.9-fold. No changes were observed in the fexofenadine elimination half-life (P > .12). The fexofenadine AUC(infinity) was increased by lopinavir/ritonavir, likely due to increased bioavailability secondary to P-glycoprotein inhibition. After repeated administration of lopinavir/ritonavir, the interaction was attenuated compared to the single-dose effect, although a net inhibitory effect was maintained. Time-dependent inhibition of P-glycoprotein by lopinavir/ritonavir should be considered when P-glycoprotein substrates are coadministered.
Collapse
|
50
|
Ebinger M, Uhr M. ABC drug transporter at the blood-brain barrier: effects on drug metabolism and drug response. Eur Arch Psychiatry Clin Neurosci 2006; 256:294-8. [PMID: 16783492 DOI: 10.1007/s00406-006-0664-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
At the blood-brain barrier (BBB) many cellular and dynamic mechanisms influence the cerebral drug metabolism and the drug response. In this review, we focus mainly on the role P-glycoprotein (P-gp) plays at the BBB. This protein is a 170-kDa ATP-dependent drug transport protein, located in the apical membrane of endothelial cells. Utilizing ATP hydrolysis as an energy source, it exports molecules which attempt to pass through the cell membrane from the outside to the inside, protecting cells from toxins and a wide range of substances. We briefly summarize some of the currently available in vivo and in vitro methods to investigate P-gp and its substrates. Hitherto, no chemical characteristic has been discovered that clearly distinguishes substrates from non-substrates of P-gp. We discuss some examples of substrates stressing the diversity of drugs and endogenous substances that relate to P-gp either as a substrate, an inhibitor, an inducer or as a combination of the above. Finally, we discuss genetic polymorphisms of the genes encoding for P-gp and their effects on drug response.
Collapse
Affiliation(s)
- Martin Ebinger
- Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany
| | | |
Collapse
|