1
|
Zhang F, Liu W, Huang J, Chen QL, Wang DD, Zou LW, Zhao YF, Zhang WD, Xu JG, Chen HZ, Ge GB. Inhibition of drug-metabolizing enzymes by Jingyin granules: implications of herb-drug interactions in antiviral therapy. Acta Pharmacol Sin 2022; 43:1072-1081. [PMID: 34183756 PMCID: PMC8237038 DOI: 10.1038/s41401-021-00697-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/12/2021] [Indexed: 02/06/2023]
Abstract
Jingyin granules, a marketed antiviral herbal medicine, have been recommended for treating H1N1 influenza A virus infection and Coronavirus disease 2019 (COVID-19) in China. To fight viral diseases in a more efficient way, Jingyin granules are frequently co-administered in clinical settings with a variety of therapeutic agents, including antiviral drugs, anti-inflammatory drugs, and other Western medicines. However, it is unclear whether Jingyin granules modulate the pharmacokinetics of Western drugs or trigger clinically significant herb-drug interactions. This study aims to assess the inhibitory potency of the herbal extract of Jingyin granules (HEJG) against human drug-metabolizing enzymes and to clarify whether HEJG can modulate the pharmacokinetic profiles of Western drug(s) in vivo. The results clearly demonstrated that HEJG dose-dependently inhibited human CES1A, CES2A, CYPs1A, 2A6, 2C8, 2C9, 2D6, and 2E1; this herbal medicine also time- and NADPH-dependently inhibited human CYP2C19 and CYP3A. In vivo tests showed that HEJG significantly increased the plasma exposure of lopinavir (a CYP3A-substrate drug) by 2.43-fold and strongly prolonged its half-life by 1.91-fold when HEJG (3 g/kg) was co-administered with lopinavir to rats. Further investigation revealed licochalcone A, licochalcone B, licochalcone C and echinatin in Radix Glycyrrhizae, as well as quercetin and kaempferol in Folium Llicis Purpureae, to be time-dependent CYP3A inhibitors. Collectively, our findings reveal that HEJG modulates the pharmacokinetics of CYP substrate-drug(s) by inactivating CYP3A, providing key information for both clinicians and patients to use herb-drug combinations for antiviral therapy in a scientific and reasonable way.
Collapse
Affiliation(s)
- Feng Zhang
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Wei Liu
- grid.412540.60000 0001 2372 7462Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Jian Huang
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai, 201203 China
| | - Qi-long Chen
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Dan-dan Wang
- SPH Xing Ling Sci. & Tech. Pharmaceutical Co., Ltd, Shanghai, 201703 China
| | - Li-wei Zou
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Yong-fang Zhao
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China ,grid.412540.60000 0001 2372 7462Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Wei-dong Zhang
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Jian-guang Xu
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Hong-zhuan Chen
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| | - Guang-bo Ge
- grid.412540.60000 0001 2372 7462Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
2
|
Ye L, Cheng L, Deng Y, Liu H, Wu X, Wang T, Chang Q, Zhang Y, Wang D, Li Z, Yang X. Herb-Drug Interaction Between Xiyanping Injection and Lopinavir/Ritonavir, Two Agents Used in COVID-19 Pharmacotherapy. Front Pharmacol 2021; 12:773126. [PMID: 34899329 PMCID: PMC8660086 DOI: 10.3389/fphar.2021.773126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
The global epidemic outbreak of the coronavirus disease 2019 (COVID-19), which exhibits high infectivity, resulted in thousands of deaths due to the lack of specific drugs. Certain traditional Chinese medicines (TCMs), such as Xiyanping injection (XYPI), have exhibited remarkable benefits against COVID-19. Although TCM combined with Western medicine is considered an effective approach for the treatment of COVID-19, the combination may result in potential herb-drug interactions in the clinical setting. The present study aims to verify the effect of XYPI on the oral pharmacokinetics of lopinavir (LPV)/ritonavir (RTV) using an in vivo rat model and in vitro incubation model of human liver microsomes. After being pretreated with an intravenous dose of XYPI (52.5 mg/kg) for one day and for seven consecutive days, the rats received an oral dose of LPV/RTV (42:10.5 mg/kg). Except for the t1/2 of LPV is significantly prolonged from 4.66 to 7.18 h (p < 0.05) after seven consecutive days pretreatment, the pretreatment resulted in only a slight change in the other pharmacokinetic parameters of LPV. However, the pharmacokinetic parameters of RTV were significantly changed after pretreatment with XYPI, particularly in treatment for seven consecutive days, the AUC0-∞ of RTV was significantly shifted from 0.69 to 2.72 h μg/mL (p < 0.05) and the CL exhibited a tendency to decrease from 2.71 L/h to 0.94 L/h (p < 0.05), and the t1/2 of RTV prolonged from 3.70 to 5.51 h (p < 0.05), in comparison with the corresponding parameters in untreated rats. After administration of XYPI, the expression of Cyp3a1 protein was no significant changed in rats. The in vitro incubation study showed XYPI noncompetitively inhibited human CYP3A4 with an apparent Ki value of 0.54 mg/ml in a time-dependent manner. Our study demonstrated that XYPI affects the pharmacokinetics of LPV/RTV by inhibiting CYP3A4 activity. On the basis of this data, patients and clinicians can take precautions to avoid potential drug-interaction risks in COVID-19 treatment.
Collapse
Affiliation(s)
- Linhu Ye
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Lei Cheng
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Yan Deng
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Hong Liu
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Xinyu Wu
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Tingting Wang
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Zhang
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Dan Wang
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Zongze Li
- Department of Pharmacy, Bijie City First People's Hospital, Bijie, China
| | - Xixiao Yang
- Department of Pharmacy, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
3
|
Zhang F, Huang J, Liu W, Wang CR, Liu YF, Tu DZ, Liang XM, Yang L, Zhang WD, Chen HZ, Ge GB. Inhibition of drug-metabolizing enzymes by Qingfei Paidu decoction: Implication of herb-drug interactions in COVID-19 pharmacotherapy. Food Chem Toxicol 2021; 149:111998. [PMID: 33476691 PMCID: PMC7816587 DOI: 10.1016/j.fct.2021.111998] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/31/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Corona Virus Disease 2019 (COVID-19) has spread all over the world and brings significantly negative effects on human health. To fight against COVID-19 in a more efficient way, drug-drug or drug-herb combinations are frequently used in clinical settings. The concomitant use of multiple medications may trigger clinically relevant drug/herb-drug interactions. This study aims to assay the inhibitory potentials of Qingfei Paidu decoction (QPD, a Chinese medicine compound formula recommended for combating COVID-19 in China) against human drug-metabolizing enzymes and to assess the pharmacokinetic interactions in vivo. The results demonstrated that QPD dose-dependently inhibited CYPs1A, 2A6, 2C8, 2C9, 2C19, 2D6 and 2E1 but inhibited CYP3A in a time- and NADPH-dependent manner. In vivo test showed that QPD prolonged the half-life of lopinavir (a CYP3A substrate-drug) by 1.40-fold and increased the AUC of lopinavir by 2.04-fold, when QPD (6 g/kg) was co-administrated with lopinavir (160 mg/kg) to rats. Further investigation revealed that Fructus Aurantii Immaturus (Zhishi) in QPD caused significant loss of CYP3A activity in NADPH-generating system. Collectively, our findings revealed that QPD potently inactivated CYP3A and significantly modulated the pharmacokinetics of CYP3A substrate-drugs, which would be very helpful for the patients and clinicians to avoid potential drug-interaction risks in COVID-19 treatment.
Collapse
Affiliation(s)
- Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Huang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai, China
| | - Wei Liu
- Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao-Ran Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yan-Fang Liu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Dong-Zhu Tu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin-Miao Liang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ling Yang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-Dong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
4
|
Kuruva CS, Gandavaram SP, Shaik TB, Chintha V, Chamarthi NR, Ghosh SK, Wudayagiri R. Synthesis, Spectral Characterization, Docking Studies and Antiviral Activity of Phosphorylated Derivatives of Lopinavir Intermediate. ChemistrySelect 2019. [DOI: 10.1002/slct.201900945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Chandra S. Kuruva
- Department of ChemistrySri Venkateswara University Tirupati 517 502 India
| | - Syam P. Gandavaram
- Department of ChemistrySri Venkateswara University Tirupati 517 502 India
| | - Thaslim B. Shaik
- Department of ChemistrySri Venkateswara University Tirupati 517 502 India
| | - Venkataramaiah Chintha
- Division of Molecular BiologyDepartment of ZoologySri Venkateswara University Tirupati 517 502 India
| | - Naga R. Chamarthi
- Department of ChemistrySri Venkateswara University Tirupati 517 502 India
| | - Sunil K. Ghosh
- Bioorganic DivisionBhabha Atomic Research Centre Mumbai 400 085 India
| | - Rajendra Wudayagiri
- Division of Molecular BiologyDepartment of ZoologySri Venkateswara University Tirupati 517 502 India
| |
Collapse
|
5
|
Nassar T, Rohald A, Naraykin N, Barasch D, Amsalem O, Prabhu P, Kotler M, Benita S. Nanocapsules embedded in microparticles for enhanced oral bioavailability and efficacy of Lopinavir as an anti-AIDS drug. J Drug Target 2018; 27:590-600. [PMID: 30470150 DOI: 10.1080/1061186x.2018.1552275] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Lopinavir (LPV), an efficient drug for HIV infection treatment, was incorporated into biodegradable PLGA nanocapsules (NCs) embedded in microparticles (MCPs) using the spray-drying technique in an attempt to bypass the P-gp efflux and protect the drug from CYP3A pre-systemic metabolism without ritonavir (RTV). SEM observations confirmed the formation of NCs and their entrapment in the MCPs. LPV-loaded NCs and free LPV were released from the MCPs at pH of 7.4 as evidenced by in vitro release studies. Results obtained from rat studies showed a two-fold higher bioavailability of LPV following oral administration of the optimal formulation than Kaletra®, the marketed drug, showing that when properly entrapped, LPV can be effectively protected from CYP degradation in the gut as well as from the liver following systemic absorption. It was also shown that serum derived from rats following LPV oral administration in two formulations and Kaletra® significantly decreased the multiplication of HIV-1 in cultured SupT1 cells. Furthermore, the LPV formulations markedly restricted the titre of infectious HIV-1 production compared with Kaletra® confirming the improved antiviral activity of LPV delivered in the rat blood circulation by the nanocapsules embedded in microparticle formulations.
Collapse
Affiliation(s)
- Taher Nassar
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Ayala Rohald
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Natalya Naraykin
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Dinorah Barasch
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Orit Amsalem
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Ponnandy Prabhu
- b Department of Pathology and Immunology , The Lautenberg Center for General and Tumor Immunology, The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Moshe Kotler
- b Department of Pathology and Immunology , The Lautenberg Center for General and Tumor Immunology, The Hebrew University of Jerusalem , Jerusalem , Israel
| | - Simon Benita
- a The Institute for Drug Research of the School of Pharmacy , The Hebrew University of Jerusalem , Jerusalem , Israel
| |
Collapse
|
6
|
Gannon PJ, Akay-Espinoza C, Yee AC, Briand LA, Erickson MA, Gelman BB, Gao Y, Haughey NJ, Zink MC, Clements JE, Kim NS, Van De Walle G, Jensen BK, Vassar R, Pierce RC, Gill AJ, Kolson DL, Diehl JA, Mankowski JL, Jordan-Sciutto KL. HIV Protease Inhibitors Alter Amyloid Precursor Protein Processing via β-Site Amyloid Precursor Protein Cleaving Enzyme-1 Translational Up-Regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:91-109. [PMID: 27993242 DOI: 10.1016/j.ajpath.2016.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 08/22/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Mounting evidence implicates antiretroviral (ARV) drugs as potential contributors to the persistence and evolution of clinical and pathological presentation of HIV-associated neurocognitive disorders in the post-ARV era. Based on their ability to induce endoplasmic reticulum (ER) stress in various cell types, we hypothesized that ARV-mediated ER stress in the central nervous system resulted in chronic dysregulation of the unfolded protein response and altered amyloid precursor protein (APP) processing. We used in vitro and in vivo models to show that HIV protease inhibitor (PI) class ARVs induced neuronal damage and ER stress, leading to PKR-like ER kinase-dependent phosphorylation of the eukaryotic translation initiation factor 2α and enhanced translation of β-site APP cleaving enzyme-1 (BACE1). In addition, PIs induced β-amyloid production, indicative of increased BACE1-mediated APP processing, in rodent neuroglial cultures and human APP-expressing Chinese hamster ovary cells. Inhibition of BACE1 activity protected against neuronal damage. Finally, ARVs administered to mice and SIV-infected macaques resulted in neuronal damage and BACE1 up-regulation in the central nervous system. These findings implicate a subset of PIs as potential mediators of neurodegeneration in HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Patrick J Gannon
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cagla Akay-Espinoza
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alan C Yee
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lisa A Briand
- Department of Psychology, Temple University, Philadelphia, Pennsylvania
| | - Michelle A Erickson
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin B Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Yan Gao
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Norman J Haughey
- Richard T. Johnson Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Janice E Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nicholas S Kim
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gabriel Van De Walle
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brigid K Jensen
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - R Christopher Pierce
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander J Gill
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dennis L Kolson
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Joseph L Mankowski
- Richard T. Johnson Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kelly L Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
7
|
Ravi PR, Vats R. Comparative pharmacokinetic evaluation of lopinavir and lopinavir-loaded solid lipid nanoparticles in hepatic impaired rat model. J Pharm Pharmacol 2017; 69:823-833. [DOI: 10.1111/jphp.12716] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/05/2017] [Indexed: 12/01/2022]
Abstract
Abstract
Objective
Drug-induced hepatotoxicity is a major cause of concern in patients receiving HIV/TB co-treatment. Lopinavir (LPV), an anti-HIV drug, shows poor plasma exposure due to hepatic first-pass metabolism. In this study, we investigated the effect of hepatotoxicity on pharmacokinetics of free LPV and LPV-loaded solid lipid nanoparticles (LPV SLNs) in male Wistar rats.
Methods
Hepatic impairment model in rats was developed by injecting CCl4 (i.p., 2 ml/kg). Comparative pharmacokinetic (n = 5) and tissue distribution studies (n = 3) were conducted following oral administration (20 mg/kg) of free LPV and LPV SLNs in normal and hepatic impaired rats. Isolated perfused liver (IPL) model (n = 3) and cycloheximide intervened lymphatic uptake studies (n = 3) were conducted to appreciate disposition pattern of LPV.
Key findings
In contrary to free LPV, pharmacokinetic results demonstrated no significant (P > 0.05) difference in drug plasma profile of LPV SLNs in normal and impaired rats. IPL model demonstrated trivial role of liver in disposition of LPV SLNs. Tissue distribution studies of SLNs showed higher (P < 0.05) LPV accumulation in lymphoidal organs. Pretreatment of cycloheximide significantly (P < 0.05) reduced AUC and Cmax of LPV SLNs.
Conclusion
From the results, we conclude that unlike conventional formulations of LPV, disposition characteristics of LPV SLNs were similar both in normal and hepatic impaired rats.
Collapse
Affiliation(s)
- Punna Rao Ravi
- Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Hyderabad, Telangana, India
| | - Rahul Vats
- Novartis Healthcare, Hyderabad, Telangana, India
| |
Collapse
|
8
|
Jensen BK, Monnerie H, Mannell MV, Gannon PJ, Espinoza CA, Erickson MA, Bruce-Keller AJ, Gelman BB, Briand LA, Pierce RC, Jordan-Sciutto KL, Grinspan JB. Altered Oligodendrocyte Maturation and Myelin Maintenance: The Role of Antiretrovirals in HIV-Associated Neurocognitive Disorders. J Neuropathol Exp Neurol 2015; 74:1093-118. [PMID: 26469251 PMCID: PMC4608376 DOI: 10.1097/nen.0000000000000255] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Despite effective viral suppression through combined antiretroviral therapy (cART), approximately half of HIV-positive individuals have HIV-associated neurocognitive disorders (HAND). Studies of antiretroviral-treated patients have revealed persistent white matter abnormalities including diffuse myelin pallor, diminished white matter tracts, and decreased myelin protein mRNAs. Loss of myelin can contribute to neurocognitive dysfunction because the myelin membrane generated by oligodendrocytes is essential for rapid signal transduction and axonal maintenance. We hypothesized that myelin changes in HAND are partly due to effects of antiretroviral drugs on oligodendrocyte survival and/or maturation. We showed that primary mouse oligodendrocyte precursor cell cultures treated with therapeutic concentrations of HIV protease inhibitors ritonavir or lopinavir displayed dose-dependent decreases in oligodendrocyte maturation; however, this effect was rapidly reversed after drug removal. Conversely, nucleoside reverse transcriptase inhibitor zidovudine had no effect. Furthermore, in vivo ritonavir administration to adult mice reduced frontal cortex myelin protein levels. Finally, prefrontal cortex tissue from HIV-positive individuals with HAND on cART showed a significant decrease in myelin basic protein compared with untreated HIV-positive individuals with HAND or HIV-negative controls. These findings demonstrate that antiretrovirals can impact myelin integrity and have implications for myelination in juvenile HIV patients and myelin maintenance in adults on lifelong therapy.
Collapse
Affiliation(s)
- Brigid K. Jensen
- Department of Neuroscience, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hubert Monnerie
- Department of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Maggie V. Mannell
- Department of Neuroscience, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patrick J. Gannon
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cagla Akay Espinoza
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michelle A. Erickson
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Annadora J. Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Benjamin B. Gelman
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Lisa A. Briand
- Department of Psychology, College of Liberal Arts, Temple University, Philadelphia, Pennsylvania
| | - R. Christopher Pierce
- Center for Neurobiology and Behavior, Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly L. Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Judith B. Grinspan
- Department of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Ravi PR, Vats R, Balija J, Adapa SPN, Aditya N. Modified pullulan nanoparticles for oral delivery of lopinavir: Formulation and pharmacokinetic evaluation. Carbohydr Polym 2014; 110:320-8. [DOI: 10.1016/j.carbpol.2014.03.099] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/16/2014] [Accepted: 03/28/2014] [Indexed: 11/28/2022]
|
10
|
Ravi PR, Vats R, Dalal V, Murthy AN. A hybrid design to optimize preparation of lopinavir loaded solid lipid nanoparticles and comparative pharmacokinetic evaluation with marketed lopinavir/ritonavir coformulation. J Pharm Pharmacol 2014; 66:912-26. [DOI: 10.1111/jphp.12217] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 12/15/2013] [Indexed: 11/26/2022]
Abstract
Abstract
Objectives
To prepare stearic acid-based lopinavir (LPV) loaded solid lipid nanoparticles (SLNs) using a hybrid design and compare in-vivo performance of optimized formulation with marketed LPV/ritonavir (RTV) coformulation.
Methods
LPV SLNs were prepared by hot melt emulsion technique and optimized using Plackett–Burman design and Box–Behnken design. Physical characterization studies were conducted for the optimized SLNs. Comparative oral pharmacokinetic studies and tissue distribution studies of optimized SLNs and LPV/RTV coformulation were done in Wistar rats. In-vitro metabolic stability and intestinal permeability studies for LPV SLNs were undertaken to elucidate the mechanism involved in the pharmacokinetic improvement of LPV.
Key findings
Optimized SLNs exhibited nanometeric size (223 nm) with high entrapment efficiency (83%). In-vitro drug release study of SLNs showed biphasic sustained release behaviour. Significant increase in oral bioavailability of LPV from LPV SLNs (5 folds) and LPV/RTV coformulation (3.7 folds) was observed as compared with free LPV. LPV SLNs showed better tissue distribution of LPV in HIV reservoirs than LPV/RTV coformulation. In-vitro studies demonstrated that SLNs provided metabolic protection of LPV and were endocytosized during absorption.
Conclusions
SLNs enhanced oral bioavailability and improved distribution profile of LPV to HIV reservoirs and hence could be better alternative to LPV/RTV coformulation.
Collapse
Affiliation(s)
- Punna Rao Ravi
- Pharmacy Department, BITS-Pilani Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Rahul Vats
- Pharmacy Department, BITS-Pilani Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | - Vikas Dalal
- Pharmacy Department, BITS-Pilani Hyderabad Campus, Hyderabad, Andhra Pradesh, India
| | | |
Collapse
|
11
|
Akay C, Cooper M, Odeleye A, Jensen BK, White MG, Vassoler F, Gannon PJ, Mankowski J, Dorsey JL, Buch AM, Cross SA, Cook DR, Peña MM, Andersen ES, Christofidou-Solomidou M, Lindl KA, Zink MC, Clements J, Pierce RC, Kolson DL, Jordan-Sciutto KL. Antiretroviral drugs induce oxidative stress and neuronal damage in the central nervous system. J Neurovirol 2014; 20:39-53. [PMID: 24420448 PMCID: PMC3928514 DOI: 10.1007/s13365-013-0227-1] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 12/10/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023]
Abstract
HIV-associated neurocognitive disorder (HAND), characterized by a wide spectrum of behavioral, cognitive, and motor dysfunctions, continues to affect approximately 50 % of HIV(+) patients despite the success of combination antiretroviral drug therapy (cART) in the periphery. Of note, potential toxicity of antiretroviral drugs in the central nervous system (CNS) remains remarkably underexplored and may contribute to the persistence of HAND in the cART era. Previous studies have shown antiretrovirals (ARVs) to be neurotoxic in the peripheral nervous system in vivo and in peripheral neurons in vitro. Alterations in lipid and protein metabolism, mitochondrial damage, and oxidative stress all play a role in peripheral ARV neurotoxicity. We hypothesized that ARVs also induce cellular stresses in the CNS, ultimately leading to neuronal damage and contributing to the changing clinical and pathological picture seen in HIV-positive patients in the cART era. In this report, we show that ARVs are neurotoxic in the CNS in both pigtail macaques and rats in vivo. Furthermore, in vitro, ARVs lead to accumulation of reactive oxygen species (ROS), and ultimately induction of neuronal damage and death. Whereas ARVs alone caused some activation of the endogenous antioxidant response in vitro, augmentation of this response by a fumaric acid ester, monomethyl fumarate (MMF), blocked ARV-induced ROS generation, and neuronal damage/death. These findings implicate oxidative stress as a contributor to the underlying mechanisms of ARV-induced neurotoxicity and will provide an access point for adjunctive therapies to complement ARV therapy and reduce neurotoxicity in this patient population.
Collapse
Affiliation(s)
- Cagla Akay
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Michael Cooper
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Akinleye Odeleye
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Brigid K. Jensen
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Michael G. White
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Fair Vassoler
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Patrick J. Gannon
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Joseph Mankowski
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Jamie L. Dorsey
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Alison M. Buch
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Stephanie A. Cross
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Denise R. Cook
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Michelle-Marie Peña
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - Emily S. Andersen
- Department of Medicine, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | | | - Kathryn A. Lindl
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| | - M. Christine Zink
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janice Clements
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - R. Christopher Pierce
- Department of Psychiatry, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Dennis L. Kolson
- Department of Neurology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Kelly L. Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, 240 S. 40th St, Rm 312 Levy Bldg, Philadelphia, PA 19104-6030 USA
| |
Collapse
|
12
|
Ravi PR, Vats R, Dalal V, Gadekar N, N A. Design, optimization and evaluation of poly-ɛ-caprolactone (PCL) based polymeric nanoparticles for oral delivery of lopinavir. Drug Dev Ind Pharm 2013; 41:131-40. [DOI: 10.3109/03639045.2013.850710] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
13
|
Ravi PR, Vats R, Thakur R, Srivani S, Aditya N. Effect of grapefruit juice and ritonavir on pharmacokinetics of lopinavir in Wistar rats. Phytother Res 2012; 26:1490-5. [PMID: 22308076 DOI: 10.1002/ptr.4593] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 11/15/2011] [Accepted: 11/23/2011] [Indexed: 11/07/2022]
Abstract
Lopinavir (LPV), a newer HIV protease inhibitor, has poor bioavailability being a substrate of both cytochrome P450 3A enzyme system (CYP3A) and permeability-glycoprotein (P-gp). Ritonavir (RTV) is a known inhibitor of both P-gp and CYP3A and is co-administered with LPV in anti-HIV therapy. Grapefruit juice (GFJ) is known to inhibit CYP3A and has conflicting effects, ranging from activation to inhibition, on P-gp. In this research work, the effects of GFJ and RTV on the pharmacokinetics of LPV were compared in rats. A mechanistic evaluation was undertaken using various in vitro and ex vivo studies to support the in vivo pharmacokinetic data. The plasma levels of LPV were found to increase significantly upon co-administration with GFJ in single dose as well as multidose pretreatment studies. Similar, but marginally higher, results were observed upon co-administration of LPV with RTV. No significant change in t(max) was observed in the various treatment groups. The apparent permeability of LPV in the ileum increased significantly after the pre-incubation with GFJ and RTV compared with no pre-incubation. The GFJ and RTV showed a significant and similar inhibitory effect on rat intestinal microsomes in the metabolism of LPV. The GFJ was equally effective as RTV in increasing the bioavailability of LPV.
Collapse
Affiliation(s)
- P R Ravi
- Pharmacy Department, BITS-Pilani Hyderabad Campus, Jawaharnagar, Ranga Reddy (District), Andhra Pradesh, India.
| | | | | | | | | |
Collapse
|