1
|
Leboffe L, di Masi A, Polticelli F, Trezza V, Ascenzi P. Structural Basis of Drug Recognition by Human Serum Albumin. Curr Med Chem 2020; 27:4907-4931. [DOI: 10.2174/0929867326666190320105316] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/12/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
Background:
Human serum albumin (HSA), the most abundant protein in plasma,
is a monomeric multi-domain macromolecule with at least nine binding sites for endogenous
and exogenous ligands. HSA displays an extraordinary ligand binding capacity as a depot and
carrier for many compounds including most acidic drugs. Consequently, HSA has the potential
to influence the pharmacokinetics and pharmacodynamics of drugs.
Objective:
In this review, the structural determinants of drug binding to the multiple sites of
HSA are analyzed and discussed in detail. Moreover, insight into the allosteric and competitive
mechanisms underpinning drug recognition, delivery, and efficacy are analyzed and discussed.
Conclusion:
As several factors can modulate drug binding to HSA (e.g., concurrent administration
of drugs competing for the same binding site, ligand binding to allosteric-coupled
clefts, genetic inherited diseases, and post-translational modifications), ligand binding to HSA
is relevant not only under physiological conditions, but also in the pharmacological therapy
management.
Collapse
Affiliation(s)
- Loris Leboffe
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Fabio Polticelli
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Viviana Trezza
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, Via della Vasca Navale 79, I- 00146 Roma, Italy
| |
Collapse
|
2
|
Rimac H, Tandarić T, Vianello R, Bojić M. Indomethacin Increases Quercetin Affinity for Human Serum Albumin: A Combined Experimental and Computational Study and Its Broader Implications. Int J Mol Sci 2020; 21:ijms21165740. [PMID: 32785199 PMCID: PMC7460863 DOI: 10.3390/ijms21165740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/17/2022] Open
Abstract
Human serum albumin (HSA) is the most abundant carrier protein in the human body. Competition for the same binding site between different ligands can lead to an increased active concentration or a faster elimination of one or both ligands. Indomethacin and quercetin both bind to the binding site located in the IIA subdomain. To determine the nature of the HSA-indomethacin-quercetin interactions, spectrofluorometric, docking, molecular dynamics studies, and quantum chemical calculations were performed. The results show that the indomethacin and quercetin binding sites do not overlap. Moreover, the presence of quercetin does not influence the binding constant and position of indomethacin in the pocket. However, binding of quercetin is much more favorable in the presence of indomethacin, with its position and interactions with HSA significantly changed. These results provide a new insight into drug-drug interactions, which can be important in situations when displacement from HSA or other proteins is undesirable or even desirable. This principle could also be used to deliberately prolong or shorten the xenobiotics' half-life in the body, depending on the desired outcomes.
Collapse
Affiliation(s)
- Hrvoje Rimac
- Department of Medicinal Chemistry, University of Zagreb Faculty of Pharmacy and Biochemistry, 10000 Zagreb, Croatia;
- Laboratory of Computational Modelling of Drugs, South Ural State University, 454008 Chelyabinsk, Russia
- Correspondence:
| | - Tana Tandarić
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (T.T.); (R.V.)
| | - Robert Vianello
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (T.T.); (R.V.)
| | - Mirza Bojić
- Department of Medicinal Chemistry, University of Zagreb Faculty of Pharmacy and Biochemistry, 10000 Zagreb, Croatia;
| |
Collapse
|
3
|
Rimac H, Dufour C, Debeljak Ž, Zorc B, Bojić M. Warfarin and Flavonoids Do Not Share the Same Binding Region in Binding to the IIA Subdomain of Human Serum Albumin. Molecules 2017; 22:molecules22071153. [PMID: 28696372 PMCID: PMC6152318 DOI: 10.3390/molecules22071153] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Accepted: 07/07/2017] [Indexed: 01/16/2023] Open
Abstract
Human serum albumin (HSA) binds a variety of xenobiotics, including flavonoids and warfarin. The binding of another ligand to the IIA binding site on HSA can cause warfarin displacement and potentially the elevation of its free concentration in blood. Studies dealing with flavonoid-induced warfarin displacement from HSA provided controversial results: estimated risk of displacement ranged from none to serious. To resolve these controversies, in vitro study of simultaneous binding of warfarin and eight different flavonoid aglycons and glycosides to HSA was carried out by fluorescence spectroscopy as well as molecular docking. Results show that warfarin and flavonoids do not share the same binding region in binding to HSA. Interactions were only observed at high warfarin concentrations not attainable under recommended dosing regimes. Docking experiments show that flavonoid aglycons and glycosides do not bind at warfarin high affinity sites, but rather to different regions within the IIA HSA subdomain. Thus, the risk of clinically significant warfarin-flavonoid interaction in binding to HSA should be regarded as negligible.
Collapse
Affiliation(s)
- Hrvoje Rimac
- Department of Medicinal Chemistry, University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, 10000 Zagreb, Croatia.
| | - Claire Dufour
- UMR408 SQPOV, Safety and Quality of Plant Products, INRA, Avignon University, 228 Route de l'Aérodrome, 84000 Avignon, France.
| | - Željko Debeljak
- Institute of Clinical Laboratory Diagnostics, Osijek University Hospital Center, Josipa Huttlera 4, 31000 Osijek, Croatia.
- Department of Pharmacology, School of Medicine, University of Osijek, Cara Hadrijana 10/E, 31000 Osijek, Croatia.
| | - Branka Zorc
- Department of Medicinal Chemistry, University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, 10000 Zagreb, Croatia.
| | - Mirza Bojić
- Department of Medicinal Chemistry, University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, 10000 Zagreb, Croatia.
| |
Collapse
|
4
|
Hochman J, Tang C, Prueksaritanont T. Drug–Drug Interactions Related to Altered Absorption and Plasma Protein Binding: Theoretical and Regulatory Considerations, and an Industry Perspective. J Pharm Sci 2015; 104:916-29. [DOI: 10.1002/jps.24306] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 01/28/2023]
|
5
|
Ascenzi P, Fanali G, Fasano M, Pallottini V, Trezza V. Clinical relevance of drug binding to plasma proteins. J Mol Struct 2014. [DOI: 10.1016/j.molstruc.2013.09.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
6
|
Vanstraelen K, Wauters J, Vercammen I, de Loor H, Maertens J, Lagrou K, Annaert P, Spriet I. Impact of hypoalbuminemia on voriconazole pharmacokinetics in critically ill adult patients. Antimicrob Agents Chemother 2014; 58:6782-9. [PMID: 25182655 PMCID: PMC4249353 DOI: 10.1128/aac.03641-14] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/23/2014] [Indexed: 01/30/2023] Open
Abstract
Setting the adequate dose for voriconazole is challenging due to its variable pharmacokinetics. We investigated the impact of hypoalbuminemia (<35 g/liter) on voriconazole pharmacokinetics in adult intensive care unit (ICU) patients treated with voriconazole (20 samples in 13 patients) as well as in plasma samples from ICU patients that had been spiked with voriconazole at concentrations of 1.5 mg/liter, 2.9 mg/liter, and 9.0 mg/liter (66 samples from 22 patients). Plasma albumin concentrations ranged from 13.8 to 38.7 g/liter. Total voriconazole concentrations in adult ICU patients treated with voriconazole ranged from 0.5 to 8.7 mg/liter. Unbound and bound voriconazole concentrations were separated using high-throughput equilibrium dialysis followed by liquid chromatography-tandem mass spectrometry (LC-MSMS). Multivariate analysis revealed a positive relationship between voriconazole plasma protein binding and plasma albumin concentrations (P < 0.001), indicating higher unbound voriconazole concentrations with decreasing albumin concentrations. The correlation is more pronounced in the presence of elevated bilirubin concentrations (P = 0.05). We therefore propose to adjust the measured total voriconazole concentrations in patients with abnormal plasma albumin and total serum bilirubin plasma concentrations who show adverse events potentially related to voriconazole via a formula that we developed. Assuming 50% protein binding on average and an upper limit of 5.5 mg/liter for total voriconazole concentrations, the upper limit for unbound voriconazole concentrations is 2.75 mg/liter. Alterations in voriconazole unbound concentrations caused by hypoalbuminemia and/or elevated bilirubin plasma concentrations cannot be countered immediately, due to the adult saturated hepatic metabolism. Consequently, increased unbound voriconazole concentrations can possibly cause adverse events, even when total voriconazole concentrations are within the reference range.
Collapse
Affiliation(s)
- Kim Vanstraelen
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, University Hospitals Leuven, Leuven, Belgium
| | - Joost Wauters
- Clinical Department of General Internal Medicine, Medical Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Ine Vercammen
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, University Hospitals Leuven, Leuven, Belgium
| | - Henriette de Loor
- Department of Microbiology and Immunology, Laboratory of Nephrology and Renal Transplantation, University Hospitals Leuven/KU Leuven, Leuven, Belgium
| | - Johan Maertens
- Clinical Department of Haematology, Acute Leukaemia and Stem Cell Transplantation Unit, University Hospitals Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Clinical Department of Laboratory Medicine, Department of Microbiology and Immunology, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Clinical Pharmacology and Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Yamasaki K, Chuang VTG, Maruyama T, Otagiri M. Albumin-drug interaction and its clinical implication. Biochim Biophys Acta Gen Subj 2013; 1830:5435-43. [PMID: 23665585 DOI: 10.1016/j.bbagen.2013.05.005] [Citation(s) in RCA: 297] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND Human serum albumin acts as a reservoir and transport protein for endogenous (e.g. fatty acids or bilirubin) and exogenous compounds (e.g. drugs or nutrients) in the blood. The binding of a drug to albumin is a major determinant of its pharmacokinetic and pharmacodynamic profile. SCOPE OF REVIEW The present review discusses recent findings regarding the nature of drug binding sites, drug-albumin binding in certain diseased states or in the presence of coadministered drugs, and the potential of utilizing albumin-drug interactions in clinical applications. MAJOR CONCLUSIONS Drug-albumin interactions appear to predominantly occur at one or two specific binding sites. The nature of these drug binding sites has been fundamentally investigated as to location, size, charge, hydrophobicity or changes that can occur under conditions such as the content of the endogenous substances in question. Such findings can be useful tools for the analysis of drug-drug interactions or protein binding in diseased states. A change in protein binding is not always a problem in terms of drug therapy, but it can be used to enhance the efficacy of therapeutic agents or to enhance the accumulation of radiopharmaceuticals to targets for diagnostic purposes. Furthermore, several extracorporeal dialysis procedures using albumin-containing dialysates have proven to be an effective tool for removing endogenous toxins or overdosed drugs from patients. GENERAL SIGNIFICANCE Recent findings related to albumin-drug interactions as described in this review are useful for providing safer and efficient therapies and diagnoses in clinical settings. This article is part of a Special Issue entitled Serum Albumin.
Collapse
Affiliation(s)
- Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | | | | | | |
Collapse
|
8
|
Human serum albumin: from bench to bedside. Mol Aspects Med 2011; 33:209-90. [PMID: 22230555 DOI: 10.1016/j.mam.2011.12.002] [Citation(s) in RCA: 1211] [Impact Index Per Article: 93.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 12/21/2011] [Indexed: 02/07/2023]
Abstract
Human serum albumin (HSA), the most abundant protein in plasma, is a monomeric multi-domain macromolecule, representing the main determinant of plasma oncotic pressure and the main modulator of fluid distribution between body compartments. HSA displays an extraordinary ligand binding capacity, providing a depot and carrier for many endogenous and exogenous compounds. Indeed, HSA represents the main carrier for fatty acids, affects pharmacokinetics of many drugs, provides the metabolic modification of some ligands, renders potential toxins harmless, accounts for most of the anti-oxidant capacity of human plasma, and displays (pseudo-)enzymatic properties. HSA is a valuable biomarker of many diseases, including cancer, rheumatoid arthritis, ischemia, post-menopausal obesity, severe acute graft-versus-host disease, and diseases that need monitoring of the glycemic control. Moreover, HSA is widely used clinically to treat several diseases, including hypovolemia, shock, burns, surgical blood loss, trauma, hemorrhage, cardiopulmonary bypass, acute respiratory distress syndrome, hemodialysis, acute liver failure, chronic liver disease, nutrition support, resuscitation, and hypoalbuminemia. Recently, biotechnological applications of HSA, including implantable biomaterials, surgical adhesives and sealants, biochromatography, ligand trapping, and fusion proteins, have been reported. Here, genetic, biochemical, biomedical, and biotechnological aspects of HSA are reviewed.
Collapse
|
9
|
Hewavitharana AK, Hyde C, Thomas R, Shaw PN. Shortcomings of protein removal prior to high performance liquid chromatographic analysis—A case study using method development for BAY 11-7082. J Chromatogr B Analyt Technol Biomed Life Sci 2006; 834:93-7. [PMID: 16530027 DOI: 10.1016/j.jchromb.2006.02.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Revised: 02/15/2006] [Accepted: 02/17/2006] [Indexed: 11/23/2022]
Abstract
During the analytical method development for BAY 11-7082 ((E)-3-[4-methylphenylsulfonyl]-2-propenenitrile), using HPLC-MS-MS and HPLC-UV, we observed that the protein removal process (both ultrafiltration and precipitation method using organic solvents) prior to HPLC brought about a significant reduction in the concentration of this compound. The use of a structurally similar internal standard, BAY 11-7085 ((E)-3-[4-t-butylphenylsulfonyl]-2-propenenitrile), was not effective in compensating for the loss of analyte as the extent of reduction was different to that of the analyte. We present here a systematic investigation of this problem and a new validated method for the determination of BAY 11-7082.
Collapse
Affiliation(s)
- A K Hewavitharana
- School of Pharmacy, University of Queensland, Brisbane, Qld 4072, Australia.
| | | | | | | |
Collapse
|
10
|
Uchiyama-Kokubu N, Watanabe T, Nakajima M. A bioassay for the activity of PSC 833 in human serum for modulation of P-glycoprotein-mediated multidrug resistance. Anticancer Drugs 2000; 11:583-90. [PMID: 11036963 DOI: 10.1097/00001813-200008000-00011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We established a rapid and sensitive ex vivo bioassay to detect the multidrug resistance (MDR)-inhibitory activity of SDZ PSC 833 ([3'-keto-Bmt1]-[Val2]-cyclosporin (PSC 833)) in two RPMI 8226 human myeloma sublines (parent 8226 and doxorubicin-resistant subline Dox6) in 75% human serum. In vitro sensitivity of the tumor to doxorubicin was determined by 3-h drug exposure growth inhibition assay (MTT assay). PSC 833 in serum restored the IC50 of doxorubicin in the P-glycoprotein (P-gp)-positive resistant subline to the same level as in the sensitive cells at 1 microg/ml, which has been shown to be an achievable concentration in clinical trials. In addition, the cytotoxic effect of doxorubicin was enhanced by PSC 833 in the sera of the patient in whom the blood level was 705.7 ng/ml. However, 10 microg/ml PSC 833 in serum does not cause a complete recovery in the IC90 of doxorubicin in the resistant sublines. This MDR-inhibitory activity was supported by the finding that PSC 833 in serum does not increase accumulation of rhodamine 123 in doxorubicin-resistant cells in an in vitro functional assay. The present study provides evidence that PSC 833 in human serum is effective to modulate P-gp-mediated MDR but insufficient for the reversal of MDR from the clinicopharmacological point of view.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Aged
- Antineoplastic Agents/pharmacology
- Biological Assay
- Cell Division
- Cyclosporins/blood
- Cyclosporins/pharmacology
- Dose-Response Relationship, Drug
- Doxorubicin/pharmacology
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Female
- Humans
- Multiple Myeloma/drug therapy
- Multiple Myeloma/enzymology
- Multiple Myeloma/pathology
- RNA, Messenger/biosynthesis
- Tumor Cells, Cultured
Collapse
|
11
|
Pacifici GM, Viani A. Methods of determining plasma and tissue binding of drugs. Pharmacokinetic consequences. Clin Pharmacokinet 1992; 23:449-68. [PMID: 1458763 DOI: 10.2165/00003088-199223060-00005] [Citation(s) in RCA: 174] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The available techniques for the investigation of drug binding to plasma and tissues protein are reviewed and the advantages and disadvantages of the various techniques stated. A comparison of different plasma protein binding techniques is made which shows that the size of the unbound fraction of drug may be influenced by the method used. Protein binding may be assayed by methods including equilibrium dialysis, ultrafiltration, ultracentrifugation, gel filtration, binding to albumin microspheres and circular dichroism. Tissue binding techniques can involve testing binding to isolated organs, tissue slices, homogenates and isolated subcellular particles. Details of the available methods to compute pharmacokinetic constants are given. Stereoselective binding has been investigated for a limited number of drugs and the difference in the binding of 2 enantiomers is usually modest. The measurement of the binding constants is often required to characterise the drug-protein interaction. Mathematical and graphical methods to compute the pharmacokinetic parameters are discussed. The implications of binding on the volume of distribution and clearance of drugs are examined.
Collapse
Affiliation(s)
- G M Pacifici
- Department of Biomedicine, University of Pisa Medical School, Italy
| | | |
Collapse
|
12
|
Tissue Binding versus Plasma Binding of Drugs: General Principles and Pharmacokinetic Consequences. ADVANCES IN DRUG RESEARCH 1991. [DOI: 10.1016/b978-0-12-013320-8.50006-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
13
|
Affiliation(s)
- J J MacKichan
- College of Pharmacy, Ohio State University, Columbus
| |
Collapse
|
14
|
Smallwood RH, Mihaly GW, Smallwood RA, Morgan DJ. Effect of a protein binding change on unbound and total plasma concentrations for drugs of intermediate hepatic extraction. JOURNAL OF PHARMACOKINETICS AND BIOPHARMACEUTICS 1988; 16:529-42. [PMID: 3199317 DOI: 10.1007/bf01062383] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
For substances eliminated from blood by the liver, the effect of a change in unbound fraction of drug (fu(b)) on steady state total (Cb) and unbound (Cu(b)) blood concentrations has hitherto only been considered for the two limiting cases, i.e., at the upper and lower extremes of hepatic intrinsic clearance (CL(int)). For a substance of very low CL(int), if fu(b) changes, Cb will change and Cub will remain constant, whereas if CL(int) is very high, Cub will change and Cb will remain constant. The present study defines the effects of a change in fu(b) on Cb and Cub over the whole CL(int) range. Computer simulations were undertaken which predicted that, for a given change in fu(b), absolute and relative changes in Cb would decrease nonlinearly with increasing CL(int), while the relative change in Cub would increase with CL(int). The absolute change in Cub would be independent of CL(int). Significant changes in Cb and Cub would be observed at intermediate values of CL(int) not just at the high and low extremes. These theoretical predictions were investigated experimentally in the isolated perfused rat liver by examining the effects of a change in fu(b) of sodium taurocholate a substance with intermediate CL(int) (such that at fu(b) = 0.27, hepatic extraction ratio = 0.71) induced by concurrent administration of sodium oleate. Sodium 24-14 C-taurocholate (specific activity 52 microCi/mmol) was infused into the reservoir in a recycling system at 30 mumol/hr for 105 min (n = 6). At 45 min a bolus dose of sodium oleate (50 mmol) was administered to the reservoir, followed by a constant infusion of 143 mmol/hr for 1 hr. Following the administration of oleate, taurocholate fu(b) fell promptly by 55% (0.27-0.12). There was a relative increase of taurocholate Cb of 22.7% and a relative decrease in Cub of 45.4%, in accordance with the simulations (p less than 0.05). We conclude that important changes in unbound steady-state concentration, the pharmacologically active moiety, can occur upon changes in unbound fraction with compounds of intermediate hepatic intrinsic clearance.
Collapse
|
15
|
|
16
|
Cayen MN. Disposition, metabolism and pharmacokinetics of antihyperlipidemic agents in laboratory animals and man. Pharmacol Ther 1985; 29:157-204. [PMID: 3915091 DOI: 10.1016/0163-7258(85)90028-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
17
|
Keller F, Maiga M, Neumayer HH, Lode H, Distler A. Pharmacokinetic effects of altered plasma protein binding of drugs in renal disease. Eur J Drug Metab Pharmacokinet 1984; 9:275-82. [PMID: 6519129 DOI: 10.1007/bf03189651] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The measurement of plasma drug concentrations provides no insight into the relationship between the free and the plasma-protein-bound fractions of drugs. Plasma protein binding may decrease in renal disease due to uremia, hypoalbuminemia, or due to drug interactions. Decreased plasma protein binding leads to an increase in free plasma fraction causing an increase in volume of distribution and a shorter elimination half life. The increase in the apparent volume of distribution and the shorter elimination half life cause a decrease in total plasma concentration. Therefore, the free drug concentration is more reliable than the total plasma concentration for therapeutic drug monitoring. However, the free amount in plasma and in tissue and the tissue-bound amount remain unchanged under steady state conditions. Thus, a decrease in plasma protein binding in renal disease usually does not lead to increased drug toxicity, and alteration of drug dosage is not required, although the total plasma concentration may be found to be considerably lower than normal. In addition to plasma protein binding, alteration of tissue binding must also be considered for the determination of the appropriate dosage of some drugs in renal disease.
Collapse
|
18
|
Abstract
The available evidence suggests that for most drugs, adverse effects in the fetus may vary with gestational state and among species due to: (i) changes in the fetal exposure to the drug (i.e. due to changes in the pharmacokinetics of the drug in the mother and/or the fetus), or to (ii) changes in the susceptibility of the fetus to the drug. The fetal exposure to a drug during gestation is influenced more by the varying capacity of mother and/or fetus to eliminate the drug than by any intrinsic 'barrier' phenomenon at the placenta. Although differential maternal/fetal protein binding, active transplacental transport processes and 'ion-trapping' effects may influence the fetal exposure of some drugs, the main mechanisms by which fetal exposure may be modulated during pregnancy are via the capacity for irreversible drug elimination--by the fetus or, less often, by the placenta. The susceptibility of a fetus to adverse drug reactions is determined by the ontogeny of vital processes and the nature of the interaction between the drug and the process. Hence 'gestational state' and 'species' dependent differences in adverse drug effects, in the presence of a constant level of exposure of drug, reflect the time dependent appearance of these processes and the differences in ontogeny of the processes among species. At present, no studies have attempted to relate the measured fetal drug exposure to the intensity of a drug response at different stages of gestation or among species. Although there is a dearth of information in this field, it is apparent that in all species the placentas of all species pose little obstruction to the passage of xenobiotics (including drugs), to the fetus. The consequence of this exposure will depend on a myriad of pharmacokinetic and pharmacodynamic considerations for a given substance in a given species. Hence the outcome cannot be predicted, but must be empirically determined. Extrapolation of findings among different drugs, species and gestational states must be undertaken with caution, recognizing the above considerations and limitations.
Collapse
|