1
|
Alsmadi MM. The investigation of the complex population-drug-drug interaction between ritonavir-boosted lopinavir and chloroquine or ivermectin using physiologically-based pharmacokinetic modeling. Drug Metab Pers Ther 2023; 38:87-105. [PMID: 36205215 DOI: 10.1515/dmpt-2022-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/19/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Therapy failure caused by complex population-drug-drug (PDDI) interactions including CYP3A4 can be predicted using mechanistic physiologically-based pharmacokinetic (PBPK) modeling. A synergy between ritonavir-boosted lopinavir (LPVr), ivermectin, and chloroquine was suggested to improve COVID-19 treatment. This work aimed to study the PDDI of the two CYP3A4 substrates (ivermectin and chloroquine) with LPVr in mild-to-moderate COVID-19 adults, geriatrics, and pregnancy populations. METHODS The PDDI of LPVr with ivermectin or chloroquine was investigated. Pearson's correlations between plasma, saliva, and lung interstitial fluid (ISF) levels were evaluated. Target site (lung epithelial lining fluid [ELF]) levels of ivermectin and chloroquine were estimated. RESULTS Upon LPVr coadministration, while the chloroquine plasma levels were reduced by 30, 40, and 20%, the ivermectin plasma levels were increased by a minimum of 425, 234, and 453% in adults, geriatrics, and pregnancy populations, respectively. The established correlation equations can be useful in therapeutic drug monitoring (TDM) and dosing regimen optimization. CONCLUSIONS Neither chloroquine nor ivermectin reached therapeutic ELF levels in the presence of LPVr despite reaching toxic ivermectin plasma levels. PBPK modeling, guided with TDM in saliva, can be advantageous to evaluate the probability of reaching therapeutic ELF levels in the presence of PDDI, especially in home-treated patients.
Collapse
Affiliation(s)
- Mo'tasem M Alsmadi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
2
|
Fayed ND, Essa EA, El Maghraby GM. Menthol augmented niosomes for enhanced intestinal absorption of lopinavir. Pharm Dev Technol 2022; 27:956-964. [PMID: 36227222 DOI: 10.1080/10837450.2022.2136195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Lopinavir is effective in treatment of HIV infection but experiences low oral bioavailability due to poor solubility, pre-systemic metabolism, and P-gp intestinal efflux. Co-processing with menthol enhanced its dissolution and intestinal permeability. Niosomes comprising Span 60, cholesterol and poloxamer 407 were formulated in absence and presence of menthol. These were evaluated for size, morphology, entrapment efficiency (EE%), lopinavir release and intestinal absorption. The later employed in situ rabbit intestinal absorption model. Niosomes were spherical with vesicle size of 140.2 ± 23 and 148.2 ± 27nm for standard and menthol containing niosomes, respectively. The EE% values were 94.4% and 96.3% for both formulations, respectively. Niosomes underwent slow release during the time course of absorption with menthol hastening lopinavir release, but the release did not exceed 9%. Niosmoal encapsulation enhanced lopinavir intestinal absorption compared with drug solution. This was reflected from the fraction absorbed from duodenum which was 24.15%, 73.09% and 83.23% for solution, standard niosomes and menthol containing vesicles, respectively. These values were 34.32%, 80.8% and 86.56% for the same formulations in case of jejuno-ileum. Lopinavir absorption from niosomes didn't depend on release supporting intact vesicle absorption. The study introduced menthol containing niosomes as carriers for enhanced lopinavir intestinal absorption.
Collapse
Affiliation(s)
- Noha D Fayed
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ebtesam A Essa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gamal M El Maghraby
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
3
|
Fayed ND, Arafa MF, Essa EA, El Maghraby GM. Lopinavir-menthol co-crystals for enhanced dissolution rate and intestinal absorption. J Drug Deliv Sci Technol 2022; 74:103587. [PMID: 35845293 PMCID: PMC9272570 DOI: 10.1016/j.jddst.2022.103587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/27/2022] [Accepted: 07/08/2022] [Indexed: 12/25/2022]
Abstract
Lopinavir is an antiretroviral, antiparasitic agent and recently utilized in treatment of COVID-19. Unfortunately, lopinavir exhibited poor oral bioavailability due to poor dissolution, extensive pre-systemic metabolism, and significant P-glycoprotein intestinal efflux. Accordingly, the aim was to enhance dissolution rate and intestinal absorption of lopinavir. This employed co-processing with menthol which is believed to modify crystalline structures and inhibit intestinal efflux. Lopinavir was mixed with menthol at different molar ratios before ethanol assisted kneading. Formulations were evaluated using FTIR spectroscopy, differential scanning calorimetry (DSC), X-ray powder diffraction (XRD) and dissolution studies. Optimum ratio was utilized to assess lopinavir intestinal permeability. This employed in situ rabbit intestinal perfusion technique. FTIR, DSC and XRD indicated formation of lopinavir-menthol co-crystals at optimum molar ratio of 1:2. Additional menthol underwent phase separation due to possible self-association. Co-crystallization significantly enhanced lopinavir dissolution rate compared with pure drug to increase the dissolution efficiency from 24.96% in case of unprocessed lopinavir to 91.43% in optimum formulation. Lopinavir showed incomplete absorption from duodenum and jejuno-iliac segments with lower absorptive clearance from jejuno-ileum reflecting P-gp efflux. Co-perfusion with menthol increased lopinavir intestinal permeability. The study introduced menthol as co-crystal co-former for enhanced dissolution and augmented intestinal absorption of lopinavir.
Collapse
|
4
|
Prodrug Therapies for Infectious and Neurodegenerative Diseases. Pharmaceutics 2022; 14:pharmaceutics14030518. [PMID: 35335894 PMCID: PMC8953076 DOI: 10.3390/pharmaceutics14030518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Prodrugs are bioreversible drug derivatives which are metabolized into a pharmacologically active drug following chemical or enzymatic modification. This approach is designed to overcome several obstacles that are faced by the parent drug in physiological conditions that include rapid drug metabolism, poor solubility, permeability, and suboptimal pharmacokinetic and pharmacodynamic profiles. These suboptimal physicochemical features can lead to rapid drug elimination, systemic toxicities, and limited drug-targeting to disease-affected tissue. Improving upon these properties can be accomplished by a prodrug design that includes the careful choosing of the promoiety, the linker, the prodrug synthesis, and targeting decorations. We now provide an overview of recent developments and applications of prodrugs for treating neurodegenerative, inflammatory, and infectious diseases. Disease interplay reflects that microbial infections and consequent inflammation affects neurodegenerative diseases and vice versa, independent of aging. Given the high prevalence, personal, social, and economic burden of both infectious and neurodegenerative disorders, therapeutic improvements are immediately needed. Prodrugs are an important, and might be said a critical tool, in providing an avenue for effective drug therapy.
Collapse
|
5
|
Scalise M, Pochini L, Galluccio M, Console L, Indiveri C. Glutamine transporters as pharmacological targets: From function to drug design. Asian J Pharm Sci 2020; 15:207-219. [PMID: 32373200 PMCID: PMC7193454 DOI: 10.1016/j.ajps.2020.02.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/18/2020] [Accepted: 02/29/2020] [Indexed: 12/17/2022] Open
Abstract
Among the different targets of administered drugs, there are membrane transporters that play also a role in drug delivery and disposition. Moreover, drug-transporter interactions are responsible for off-target effects of drugs underlying their toxicity. The improvement of the drug design process is subjected to the identification of those membrane transporters mostly relevant for drug absorption, delivery and side effect production. A peculiar group of proteins with great relevance to pharmacology is constituted by the membrane transporters responsible for managing glutamine traffic in different body districts. The interest around glutamine metabolism lies in its physio-pathological role; glutamine is considered a conditionally essential amino acid because highly proliferative cells have an increased request of glutamine that cannot be satisfied only by endogenous synthesis. Then, glutamine transporters provide cells with this special nutrient. Among the glutamine transporters, SLC1A5, SLC6A14, SLC6A19, SLC7A5, SLC7A8 and some members of SLC38 family are the best characterized, so far, in both physiological and pathological conditions. Few 3D structures have been solved by CryoEM; other structural data on these transporters have been obtained by computational analysis. Interactions with drugs have been described for several transporters of this group. For some of them, the studies are at an advanced stage, for others, the studies are still in nuce and novel biochemical findings open intriguing perspectives.
Collapse
Affiliation(s)
- Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lorena Pochini
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Lara Console
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| |
Collapse
|
6
|
Saeheng T, Na-Bangchang K, Siccardi M, Rajoli RKR, Karbwang J. Physiologically-Based Pharmacokinetic Modeling for Optimal Dosage Prediction of Quinine Coadministered With Ritonavir-Boosted Lopinavir. Clin Pharmacol Ther 2020; 107:1209-1220. [PMID: 31721171 DOI: 10.1002/cpt.1721] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/03/2019] [Indexed: 12/25/2022]
Abstract
The coformulated lopinavir/ritonavir significantly reduces quinine concentration in healthy volunteers due to potential drug-drug interactions (DDIs). However, DDI information in malaria and HIV coinfected patients are lacking. The objective of the study was to apply physiologically-based pharmacokinetic (PBPK) modeling to predict optimal dosage regimens of quinine when coadministered with lopinavir/ritonavir in malaria and HIV coinfected patients with different conditions. The developed model was validated against literature. Model verification was evaluated using the accepted method. The verified PBPK models successfully predicted unbound quinine disposition when coadministered with lopinavir/ritonavir in coinfected patients with different conditions. Suitable dose adjustments to counteract with the DDIs have identified in patients with various situations (i.e., a 7-day course at 1,800 mg t.i.d. in patients with malaria with HIV infection, 648 mg b.i.d. in chronic renal failure, 648 mg t.i.d. in hepatic insufficiency except for severe hepatic insufficiency (324 mg b.i.d.), and 648 mg t.i.d. in CYP3A4 polymorphism).
Collapse
Affiliation(s)
- Teerachat Saeheng
- Leading Program, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Clinical Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Kesara Na-Bangchang
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College, Thammasat University, Pathumthani, Thailand.,Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University, Klongluang, Thailand
| | - Marco Siccardi
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Rajith K R Rajoli
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK
| | - Juntra Karbwang
- Department of Clinical Product Development, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Chulabhorn International College, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
7
|
A. M. Subbaiah M, Mandlekar S, Desikan S, Ramar T, Subramani L, Annadurai M, Desai SD, Sinha S, Jenkins SM, Krystal MR, Subramanian M, Sridhar S, Padmanabhan S, Bhutani P, Arla R, Singh S, Sinha J, Thakur M, Kadow JF, Meanwell NA. Design, Synthesis, and Pharmacokinetic Evaluation of Phosphate and Amino Acid Ester Prodrugs for Improving the Oral Bioavailability of the HIV-1 Protease Inhibitor Atazanavir. J Med Chem 2019; 62:3553-3574. [DOI: 10.1021/acs.jmedchem.9b00002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
8
|
Gilman TL, George CM, Vitela M, Herrera-Rosales M, Basiouny MS, Koek W, Daws LC. Constitutive plasma membrane monoamine transporter (PMAT, Slc29a4) deficiency subtly affects anxiety-like and coping behaviours. Eur J Neurosci 2018; 48:10.1111/ejn.13968. [PMID: 29797618 PMCID: PMC6252160 DOI: 10.1111/ejn.13968] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/04/2018] [Accepted: 05/14/2018] [Indexed: 12/27/2022]
Abstract
Originally, uptake-mediated termination of monoamine (e.g., serotonin and dopamine) signalling was believed to only occur via high-affinity, low-capacity transporters ("uptake1 ") such as the serotonin or dopamine transporters, respectively. Now, the important contribution of a second low-affinity, high-capacity class of biogenic amine transporters has been recognised, particularly in circumstances when uptake1 transporter function is reduced (e.g., antidepressant treatment). Pharmacologic or genetic reductions in uptake1 function can change locomotor, anxiety-like or stress-coping behaviours. Comparable behavioural investigations into reduced low-affinity, high-capacity transporter function are lacking, in part, due to a current dearth of drugs that selectively target particular low-affinity, high-capacity transporters, such as the plasma membrane monoamine transporter. Therefore, the most direct approach involves constitutive genetic knockout of these transporters. Other groups have reported that knockout of the low-affinity, high-capacity organic cation transporters 2 or 3 alters anxiety-like and stress-coping behaviours, but none have assessed behaviours in plasma membrane monoamine transporter knockout mice. Here, we evaluated adult male and female plasma membrane monoamine transporter wild-type, heterozygous and knockout mice in locomotor, anxiety-like and stress-coping behavioural tests. A mild enhancement of anxiety-related behaviour was noted in heterozygous mice. Active-coping behaviour was modestly and selectively increased in female knockout mice. These subtle behavioural changes support a supplemental role of plasma membrane monoamine transporter in serotonin and dopamine uptake, and suggest sex differences in transporter function should be examined more closely in future investigations.
Collapse
Affiliation(s)
- T. Lee Gilman
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Christina M. George
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Melissa Vitela
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Myrna Herrera-Rosales
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mohamed S. Basiouny
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Wouter Koek
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lynette C. Daws
- Department of Cellular & Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
Subbaiah MAM, Meanwell NA, Kadow JF, Subramani L, Annadurai M, Ramar T, Desai SD, Sinha S, Subramanian M, Mandlekar S, Sridhar S, Padmanabhan S, Bhutani P, Arla R, Jenkins SM, Krystal MR, Wang C, Sarabu R. Coupling of an Acyl Migration Prodrug Strategy with Bio-activation To Improve Oral Delivery of the HIV-1 Protease Inhibitor Atazanavir. J Med Chem 2018; 61:4176-4188. [PMID: 29693401 DOI: 10.1021/acs.jmedchem.8b00277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
HIV-1 protease inhibitors (PIs), which include atazanavir (ATV, 1), remain important medicines to treat HIV-1 infection. However, they are characterized by poor oral bioavailability and a need for boosting with a pharmacokinetic enhancer, which results in additional drug-drug interactions that are sometimes difficult to manage. We investigated a chemo-activated, acyl migration-based prodrug design approach to improve the pharmacokinetic profile of 1 but failed to obtain improved oral bioavailability over dosing the parent drug in rats. This strategy was refined by conjugating the amine with a promoiety designed to undergo bio-activation, as a means of modulating the subsequent chemo-activation. This culminated in a lead prodrug that (1) yielded substantially better oral drug delivery of 1 when compared to the parent itself, the simple acyl migration-based prodrug, and the corresponding simple l-Val prodrug, (2) acted as a depot which resulted in a sustained release of the parent drug in vivo, and (3) offered the benefit of mitigating the pH-dependent absorption associated with 1, thereby potentially reducing the risk of decreased bioavailability with concurrent use of stomach-acid-reducing drugs.
Collapse
|
10
|
Subbaiah MAM, Meanwell NA, Kadow JF. Design strategies in the prodrugs of HIV-1 protease inhibitors to improve the pharmaceutical properties. Eur J Med Chem 2017; 139:865-883. [PMID: 28865281 DOI: 10.1016/j.ejmech.2017.07.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/18/2017] [Accepted: 07/21/2017] [Indexed: 11/26/2022]
Abstract
Combination antiretroviral therapy (cART) is currently the most effective treatment for HIV-1 infection. HIV-1 protease inhibitors (PIs) are an important component of some regimens of cART. However, PIs are known for sub-optimal ADME properties, resulting in poor oral bioavailability. This often necessitates high drug doses, combination with pharmacokinetic enhancers and/or special formulations in order to effectively deliver PIs, which may lead to a high pill burden and reduced patient compliance. As a remedy, improving the ADME properties of existing drugs via prodrug and other approaches has been pursued in addition to the development of next generation PIs with improved pharmacokinetic, resistance and side effect profiles. Phosphate prodrugs have been explored to address the solubility-limiting absorption and high excipient load. Prodrug design to target carrier-mediated drug delivery has also been explored. Amino acid prodrugs have been shown to improve permeability by engaging active transport mechanisms, reduce efflux and mitigate first pass metabolism while acyl migration prodrugs have been shown to improve solubility. Prodrug design efforts have led to the identification of one marketed agent, fosamprenavir, and clinical studies with two other prodrugs. Several of the reported approaches lack detailed in vivo characterization and hence the potential preclinical or clinical benefits of these approaches are yet to be fully determined.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Prodrug Group, Department of Medicinal Chemistry, Biocon Bristol-Myers Squibb R&D Centre, Biocon Park, Bommasandra Phase IV, Jigani Link Road, Bangalore 560009, India.
| | - Nicholas A Meanwell
- Department of Discovery Chemistry and Molecular Technologies, Bristol-Myers Squibb Research and Development, PO Box 4000, Princeton, NJ, 08543-4000, USA
| | - John F Kadow
- Department of Medicinal Chemistry, ViiV Healthcare, 36 East Industrial Road, Branford, CT 06405, USA
| |
Collapse
|
11
|
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
|
12
|
Murakami T. A Minireview: Usefulness of Transporter-Targeted Prodrugs in Enhancing Membrane Permeability. J Pharm Sci 2016; 105:2515-2526. [DOI: 10.1016/j.xphs.2016.05.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/28/2016] [Accepted: 05/03/2016] [Indexed: 11/26/2022]
|
13
|
Sutera FM, De Caro V, Giannola LI. Small endogenous molecules as moiety to improve targeting of CNS drugs. Expert Opin Drug Deliv 2016; 14:93-107. [DOI: 10.1080/17425247.2016.1208651] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Flavia Maria Sutera
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Viviana De Caro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| | - Libero Italo Giannola
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
14
|
Sheng Y, Yang X, Wang Z, Mitra AK. Stereoisomeric Prodrugs to Improve Corneal Absorption of Prednisolone: Synthesis and In Vitro Evaluation. AAPS PharmSciTech 2016; 17:718-26. [PMID: 26335418 DOI: 10.1208/s12249-015-0400-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/19/2015] [Indexed: 01/31/2023] Open
Abstract
A series of stereoisomeric prodrugs have been designed to examine efficacy in generating higher corneal absorption relative to prednisolone. Prodrugs have been studied and identified with LC/MS/MS and NMR analyses. Prodrugs have been characterized for aqueous solubility, buffer stability, and cytotoxicity. Cellular uptake and permeability studies have been conducted across MDCK-MDR1 cells to determine prodrug affinity towards P-glycoprotein (P-gp) and peptide transporters. Enzyme-mediated degradation of prodrugs has been determined using Statens Seruminstitut rabbit cornea (SIRC) cell homogenates. Prodrugs exhibited higher aqueous solubility relative to prednisolone. Prodrugs circumvented P-gp-mediated cellular efflux and were recognized by peptide transporters. Prodrugs (DP, DDP) produced with D-isomers (D-valine) were significantly stable against both chemical and enzymatic hydrolyses. The order of degradation rate constants observed in chemical and enzymatic hydrolyses were in the same order, i.e., L-valine-L-valine-prednisolone (LLP) > L-valine-D-valine-prednisolone (LDP) > D-valine-L-valine-prednisolone (DLP) > D-valine-D-valine-prednisolone (DDP). Results obtained from this study clearly suggest that stereoisomeric prodrug approach is an effective strategy to overcome P-gp-mediated efflux and improve transcorneal permeability of prednisolone following topical administration.
Collapse
|
15
|
Sheng Y, Yang X, Pal D, Mitra AK. Prodrug approach to improve absorption of prednisolone. Int J Pharm 2015; 487:242-9. [PMID: 25888804 DOI: 10.1016/j.ijpharm.2015.04.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 04/02/2015] [Accepted: 04/12/2015] [Indexed: 01/11/2023]
Abstract
Amino acid and dipeptide prodrugs have been developed to examine their potential in enhancing aqueous solubility and permeability as well as to bypass P-glycoprotein (P-gp) mediated cellular efflux of prednisolone. Prodrugs have been synthesized and identified with LC/MS/MS and NMR. Prodrugs displayed significantly higher aqueous solubility relative to prednisolone. These compounds also exhibited higher stability under acidic conditions relative to basic medium. [14]-Erythromycin uptake remained unaltered in the presence of valine-valine-prednisolone (VVP) indicating lower affinity toward P-gp. Moreover, VVP generated significantly higher transepithelial permeability across MDCK-MDR1 cells compared to prednisolone. Importantly, [3H]-GlySar uptake diminished significantly in the presence of VVP indicating high affinity toward peptide transporters. Moreover, prednisolone was regenerated from VVP due to enzymatic hydrolysis in SIRC cell homogenate. Results obtained from these studies clearly suggest that peptide transporter targeted prodrugs is a viable strategy to improve aqueous solubility and overcome P-gp mediated cellular efflux of prednisolone.
Collapse
Affiliation(s)
- Ye Sheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Xiaoyan Yang
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Dhananjay Pal
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Ashim K Mitra
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
16
|
Patel M, Sheng Y, Mandava NK, Pal D, Mitra AK. Dipeptide prodrug approach to evade efflux pumps and CYP3A4 metabolism of lopinavir. Int J Pharm 2014; 476:99-107. [PMID: 25261710 PMCID: PMC4344907 DOI: 10.1016/j.ijpharm.2014.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 09/21/2014] [Accepted: 09/23/2014] [Indexed: 12/13/2022]
Abstract
Oral absorption of lopinavir (LPV) is limited due to P-glycoprotein (P-gp) and multidrug resistance-associated protein2 (MRP2) mediated efflux by intestinal epithelial cells. Moreover, LPV is extensively metabolized by CYP3A4 enzymes. In the present study, dipeptide prodrug approach was employed to circumvent efflux pumps (P-gp and MRP2) and CYP3A4 mediated metabolism of LPV. Valine-isoleucine-LPV (Val-Ile-LPV) was synthesized and identified by LCMS and NMR techniques. The extent of LPV and Val-Ile-LPV interactions with P-gp and MRP2 was studied by uptake and transport studies across MDCK-MDR1 and MDCK-MRP2 cells. To determine the metabolic stability, time and concentration dependent degradation study was performed in liver microsomes. Val-Ile-LPV exhibited significantly higher aqueous solubility relative to LPV. This prodrug generated higher stability under acidic pH. Val-Ile-LPV demonstrated significantly lower affinity toward P-gp and MRP2 relative to LPV. Transepithelial transport of Val-Ile-LPV was significantly higher in the absorptive direction (apical to basolateral) relative to LPV. Importantly, Val-Ile-LPV was recognized as an excellent substrate by peptide transporter. Moreover, Val-Ile-LPV displayed significantly higher metabolic stability relative to LPV. Results obtained from this study suggested that dipeptide prodrug approach is a viable option to elevate systemic levels of LPV following oral administration.
Collapse
Affiliation(s)
- Mitesh Patel
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Ye Sheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Nanda K Mandava
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Dhananjay Pal
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Ashim K Mitra
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA.
| |
Collapse
|
17
|
Dahan A, Zimmermann EM, Ben-Shabat S. Modern prodrug design for targeted oral drug delivery. Molecules 2014; 19:16489-505. [PMID: 25317578 PMCID: PMC6271014 DOI: 10.3390/molecules191016489] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/07/2014] [Accepted: 10/08/2014] [Indexed: 12/20/2022] Open
Abstract
The molecular information that became available over the past two decades significantly influenced the field of drug design and delivery at large, and the prodrug approach in particular. While the traditional prodrug approach was aimed at altering various physiochemical parameters, e.g., lipophilicity and charge state, the modern approach to prodrug design considers molecular/cellular factors, e.g., membrane influx/efflux transporters and cellular protein expression and distribution. This novel targeted-prodrug approach is aimed to exploit carrier-mediated transport for enhanced intestinal permeability, as well as specific enzymes to promote activation of the prodrug and liberation of the free parent drug. The purpose of this article is to provide a concise overview of this modern prodrug approach, with useful successful examples for its utilization. In the past the prodrug approach used to be viewed as a last option strategy, after all other possible solutions were exhausted; nowadays this is no longer the case, and in fact, the prodrug approach should be considered already in the very earliest development stages. Indeed, the prodrug approach becomes more and more popular and successful. A mechanistic prodrug design that aims to allow intestinal permeability by specific transporters, as well as activation by specific enzymes, may greatly improve the prodrug efficiency, and allow for novel oral treatment options.
Collapse
Affiliation(s)
- Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | - Ellen M Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32608, USA
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|