1
|
Yanaka S, Yogo R, Yagi H, Onitsuka M, Wakaizumi N, Yamaguchi Y, Uchiyama S, Kato K. Negative interference with antibody-dependent cellular cytotoxicity mediated by rituximab from its interactions with human serum proteins. Front Immunol 2023; 14:1090898. [PMID: 36761774 PMCID: PMC9905677 DOI: 10.3389/fimmu.2023.1090898] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/03/2023] [Indexed: 01/26/2023] Open
Abstract
Although interactions of small molecular drugs with serum proteins have been widely studied from pharmacokinetic and pharmacodynamic perspectives, there have been few reports on the effects of serum components on therapeutic antibody functions. This study reports the effect of abundant serum proteins on antibody-dependent cellular cytotoxicity (ADCC) mediated by rituximab and Fcγ receptor III (FcγRIII). Human serum albumin (HSA) and the Fab fragment from the pooled serum polyclonal IgG were found to compromise ADCC as non-competitive inhibitors. Our nuclear magnetic resonance data provided direct evidence for the interactions of HSA with both the Fab and Fc regions of rituximab and also with the extracellular region of FcγRIII (sFcγRIII). The degree of involvement in the interaction decreased in the order of rituximab-Fab > rituximab-Fc > sFcγRIII, suggesting preferential binding of HSA to net positively charged proteins. Although much less pronounced than the effect of HSA, polyclonal IgG-Fab specifically interacted with rituximab-Fc. The NMR data also showed that the serum protein interactions cover the Fc surface extensively, suggesting that they can act as pan-inhibitors against various Fc receptor-mediated functions and pharmacokinetics. Our findings highlight the importance of considering serum-protein interactions in the design and application of antibody-based drugs with increased efficacy and safety.
Collapse
Affiliation(s)
- Saeko Yanaka
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Rina Yogo
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Hirokazu Yagi
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Masayoshi Onitsuka
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | | | - Yuki Yamaguchi
- Graduate School of Engineering, University of Osaka, Osaka, Japan
| | - Susumu Uchiyama
- Graduate School of Engineering, University of Osaka, Osaka, Japan
| | - Koichi Kato
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
2
|
Kerep R, Šeba T, Gabričević M. Effect of the sialic acid residues upon the binding of beta blocker propranolol to human serum alpha-1 acid glycoprotein. MAKEDONSKO FARMACEVTSKI BILTEN 2022. [DOI: 10.33320/maced.pharm.bull.2022.68.03.194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Robert Kerep
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, HR-10000 Zagreb, Croatia
| | - Tino Šeba
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, HR-10000 Zagreb, Croatia
| | - Mario Gabričević
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Ante Kovačića 1, HR-10000 Zagreb, Croatia
| |
Collapse
|
3
|
Kamaly N, Farokhzad OC, Corbo C. Nanoparticle protein corona evolution: from biological impact to biomarker discovery. NANOSCALE 2022; 14:1606-1620. [PMID: 35076049 DOI: 10.1039/d1nr06580g] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nanoparticles exposed to biological fluids such as blood, quickly interact with their surrounding milieu resulting in a biological coating that results in large part as a function of the physicochemical properties of the nanomaterial. The large nanoparticle surface area-to-volume ratio further augments binding of biological molecules and the resulting biomolecular or protein corona, once thought of as problematic biofouling, is now viewed as a rich source of biological information that can guide the development of nanomedicines. This review gives an overview of the utility of the protein corona in proteomic profiling and discusses how a better understanding of nano-bio interactions can accelerate the clinical translation of nanomedicines and facilitate the identification of disease-specific biomarkers. With the FDA requirement of the protein corona analysis of nanoparticles in place, it is envisaged that analyzing the protein corona of nanoparticles on a case-by-case basis can provide highly valuable nano-bio interface information that can aid and improve their clinical translation.
Collapse
Affiliation(s)
- Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ London, UK.
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, USA.
| | - Claudia Corbo
- Department of Medicine and Surgery, Center for Nanomedicine NANOMIB, University of Milan Bicocca, Milan, Italy.
- IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| |
Collapse
|
4
|
Ali A, Hasan P, Irfan M, Uddin A, Khan A, Saraswat J, Maguire R, Kavanagh K, Patel R, Joshi MC, Azam A, Mohsin M, Haque QMR, Abid M. Development of Oxadiazole-Sulfonamide-Based Compounds as Potential Antibacterial Agents. ACS OMEGA 2021; 6:27798-27813. [PMID: 34722980 PMCID: PMC8552329 DOI: 10.1021/acsomega.1c03379] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/30/2021] [Indexed: 06/13/2023]
Abstract
In this work, substituted 1,2,4-oxadiazoles (OX1-OX27) were screened against five bacterial strains, identified to be OX7 and OX11 as growth inhibitors with minimum inhibitory concentration (MIC) values of 31.25 and 15.75 μg/mL, respectively. The growth inhibitory property of OX7 and OX11 was further validated by disk diffusion, growth curve, and time kill curve assays. Both disrupted biofilm formation with 92-100% reduction examined by the XTT assay were further visualized by scanning electron microscopy analysis. These compounds in combination with ciprofloxacin also exhibit synergy against Escherichia coli cells. With insignificant cytotoxic behavior on HEK293 cells, human red blood cells, and Galleria mellonella larvae, OX11 was tested against 28 multidrug resistant environmental isolates of bacteria and showed inhibition of Kluyvera georgiana and Citrobacter werkmanii strains with 32 and 16 μg/mL MIC values, respectively. The synergistic behavior of OX11 with ampicillin showed many fold reductions in MIC values against K. georgiana and Klebsiella pneumoniae multidrug resistant strains. Further, transmission electron microscopy analysis of OX11-treated E. coli cells showed a significantly damaged cell wall, which resulted in the loss of integrity and cytosolic oozing. OX11 showed significant changes in the secondary structure of human serum albumin (HSA) in the presence of OX11, enhancing HSA stability. Overall, the study provided a suitable core for further synthetic alterations and development as an antibacterial agent.
Collapse
Affiliation(s)
- Asghar Ali
- Microbiology
Research Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Phool Hasan
- Medicinal
Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Irfan
- Medicinal
Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Amad Uddin
- Medicinal
Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ashba Khan
- Medicinal
Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Juhi Saraswat
- Biophysical
Chemistry Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ronan Maguire
- Department
of Biology, Maynooth University, Maynooth, Co. Kildare ABC127 Ireland
| | - Kevin Kavanagh
- Department
of Biology, Maynooth University, Maynooth, Co. Kildare ABC127 Ireland
| | - Rajan Patel
- Biophysical
Chemistry Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mukesh C. Joshi
- Motilal
Nehru College, University of Delhi, Benito Juarez Marg, South Campus, New Delhi 110021, India
| | - Amir Azam
- Department
of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohd. Mohsin
- Metabolic
Engineering Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Qazi Mohd. Rizwanul Haque
- Microbiology
Research Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Abid
- Medicinal
Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
5
|
Characterization of drug binding with alpha 1-acid glycoprotein in clinical samples using ultrafast affinity extraction. J Chromatogr A 2021; 1649:462240. [PMID: 34034105 DOI: 10.1016/j.chroma.2021.462240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 11/24/2022]
Abstract
Many drugs bind to serum transport proteins, which can affect both drug distribution and activity in the body. α1-Acid glycoprotein (AGP) is a key transport protein for basic and neutral drugs. Both elevated levels and altered glycosylation patterns of AGP have been seen in clinical conditions such as systemic lupus erythematosus (SLE). This study developed, optimized, and used the method of ultrafast affinity extraction (UAE) to examine whether these changes in AGP are associated with changes in the binding by some drugs to this transport protein. This approach used affinity microcolumns to capture and measure, in serum, the free fractions of several drugs known to bind AGP. These measurements were made with pooled normal control serum and serum samples from individuals with SLE. Immunoaffinity chromatography was used to obtain the content of AGP and HSA in these samples, and CE was used to examine the glycoform pattern for AGP in each serum sample. The free drug fractions measured for normal control serum ranged from 3.5 to 29.1%, in agreement with the results of ultrafiltration, and provided binding constants of ~105-106 M-1 for the given drugs with AGP at 37⁰C. Analysis of a screening set of SLE serum samples by UAE gave decreased free fractions (relative change, 12-55%) vs normal serum when spiked with the same types and amounts of drugs. These changes were related in some cases to AGP content, with some SLE samples having AGP levels 1.3- to 2.1-fold above the upper end of the normal range. In other cases, the changes in free fractions appeared to be linked to alterations in the glycoforms and binding constants of AGP, with some affinities differing by 1.2- to 1.5-fold vs normal AGP. This approach can be employed with other solute-protein systems and to investigate binding by other drugs or transport proteins directly in clinical samples.
Collapse
|
6
|
Severe hypoglycaemia under abemaciclib administration in a patient with breast cancer: A case report. Mol Clin Oncol 2021; 14:61. [PMID: 33604051 PMCID: PMC7849062 DOI: 10.3892/mco.2021.2223] [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: 02/12/2020] [Accepted: 01/11/2021] [Indexed: 01/22/2023] Open
Abstract
The current study reports the case of an 80-year-old woman who experienced severe hypoglycaemia after abemaciclib administration, with a recovery time of ~46 h. Abemaciclib is a cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitor that is used to treat metastatic breast cancer. A side effect of abemaciclib administration is an increase in creatinine levels. The half-life (t1/2) of 150 mg abemaciclib in patients with breast cancer was reported to be 17.5 h (nearly lower limit), and the time to reach Cmax was ~5 h (Tmax, 4-6 h). Therefore, the total time to reach half the maximum blood concentration after abemaciclib administration is ~24 h (Tmax + t1/2=5+17.5=22.5 h). As abemaciclib is administered twice daily, a considerable amount (Cmax = 123 ng/ml) may persist in the blood following the initial dose. Upon repeated administration, the blood abemaciclib concentration in patients with metastatic liver tumours might increase, although their liver function remains normal. The patient described in the current study had a creatinine level of 1.05 mg/dl at the start of abemaciclib administration. At the time of emergency hospitalisation (on day 5 of abemaciclib administration), the creatinine level was 1.40 mg/dl; however, dehydration was not observed. The patient had been administered the same dose of glimepiride for >1 year and had not experienced hypoglycaemia previously. It can be speculated that the increase in blood creatinine level had some effect on glimepiride metabolism. It is thought that administered abemaciclib enhances metabolic delay in the blood in the same way as in patients with impaired liver function, and as a result, the creatinine level increases in patients with liver metastases. This causes a decrease in renal function, which in turn results in an increase in blood concentration of glimepiride, consequently leading to severe hypoglycaemia. Therefore, clinicians must be careful when using abemaciclib in patients with liver metastases, diabetes and poor renal function.
Collapse
|
7
|
Ricciardi L, Guzzi R, Rizzuti B, Ionescu A, Aiello I, Ghedini M, La Deda M. Anionic versus neutral Pt (II) complexes: The relevance of the charge for human serum albumin binding. J Inorg Biochem 2020; 206:111024. [PMID: 32070915 DOI: 10.1016/j.jinorgbio.2020.111024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/30/2020] [Accepted: 02/09/2020] [Indexed: 10/25/2022]
Abstract
The focus of this work is pointing out the different behavior of two structurally related Pt(II) complexes, the anionic cyclometalated NBu4[(Bzq)Pt(Thio)], 1 and the neutral [(Phen)Pt(Thio)], 2, (Bzq = benzo[h]quinoline, Phen = 1,10-phenantroline, Thio = 1,2-benzenedithiolate), on the interaction with human serum albumin (HSA), a key drug-delivery protein in the bloodstream. Being very limited the number of anionic Pt(II) complexes reported to date, this is a pioneering example of report on a protein-ligand interaction involving a negatively charged platinum compound. The study was carried out by using fluorescence spectroscopy, differential scanning calorimetry and molecular docking simulations. The results revealed a strong binding affinity between the anionic compound and the protein, whereas a weak/moderate binding interaction was highlighted for the neutral one. Comparative studies with site specific ligands (warfarin and ibuprofen), allowed us to identify the protein binding sites of the two compounds. The work aims to shed light on the relevance of the charge in designing new drugs with a favorable binding affinity for HSA, which strongly contributes to influence their pharmacological and toxicological profile.
Collapse
Affiliation(s)
- Loredana Ricciardi
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy.
| | - Rita Guzzi
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy; Department of Physics, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Bruno Rizzuti
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy
| | - Andreea Ionescu
- MAT_InLAB, Department of Chemistry and Chemical Technology, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Iolinda Aiello
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy; MAT_InLAB, Department of Chemistry and Chemical Technology, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Mauro Ghedini
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy; MAT_InLAB, Department of Chemistry and Chemical Technology, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Massimo La Deda
- CNR NANOTEC - Institute of Nanotechnology, UOS Cosenza, 87036 Arcavacata di Rende, CS, Italy; MAT_InLAB, Department of Chemistry and Chemical Technology, University of Calabria, 87036 Arcavacata di Rende, CS, Italy.
| |
Collapse
|
8
|
Albumin Modifies Responses to Hematopoietic Stem Cell Mobilizing Agents in Mice. Cells 2019; 9:cells9010004. [PMID: 31861319 PMCID: PMC7017167 DOI: 10.3390/cells9010004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/13/2019] [Accepted: 12/15/2019] [Indexed: 12/15/2022] Open
Abstract
Albumin, the most abundant plasma protein, not only controls osmotic blood pressure, but also serves as a carrier for various small molecules, including pharmaceuticals. Its impact on pharmacological properties of many drugs has been extensively studied over decades. Here, we focus on its interaction with the following mobilizing agents: Granulocyte-colony stimulating factor (G-CSF) and AMD3100, where such analyses are lacking. These compounds are widely used for hematopoietic stem cell mobilization of healthy donors or patients. Using albumin-deficient (Alb−/−) mice, we studied the contribution of albumin to mobilization outcomes. Mobilization with the bicyclam CXCR4 antagonist AMD3100 was attenuated in Alb−/− mice compared to wild-type littermates. By contrast, mobilization with recombinant human G-CSF (rhG-CSF), administered twice daily over a five-day course, was significantly increased in Alb−/− mice. In terms of a mechanism, we show that rhG-CSF bioavailability in the bone marrow is significantly improved in Alb−/− mice, compared to wild-type (WT) littermates, where rhG-CSF levels dramatically drop within a few hours of the injection. These observations likely explain the favorable mobilization outcomes with split-dose versus single-dose administration of rhG-CSF to healthy donors.
Collapse
|
9
|
Nishi K, Sakurama K, Kobashigawa Y, Morioka H, Udo N, Hashimoto M, Imoto S, Yamasaki K, Otagiri M. Interaction of Aripiprazole With Human α 1-Acid Glycoprotein. J Pharm Sci 2019; 108:3911-3916. [PMID: 31520646 DOI: 10.1016/j.xphs.2019.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/25/2019] [Accepted: 09/06/2019] [Indexed: 01/30/2023]
Abstract
We recently reported that aripiprazole binds strongly to human albumin. In continuing our investigations, we investigated the mechanism responsible for the binding and the related interactions of aripiprazole with α1-acid glycoprotein (AGP). The extrinsic Cotton effects for the binding of aripiprazole and its derivatives to AGP were generated, but the magnitudes of the induced circular dichroism intensities did not correlate with those for the binding affinities. It therefore appears that the binding mode of aripiprazole with AGP is somewhat complicated, compared with that of albumin. Isothermal titration calorimetry data obtained for the binding of aripiprazole with AGP were different from that for albumin systems in that the 3 driving reactions, entropy-driven, enthalpy-driven, and the entropy-enthalpy mixed type, were all found for the AGP system, but not albumin. Moreover, the weak binding mode of aripiprazole with the 2 proteins were supported by a molecular docking model analysis. The concentration of albumin in plasma is about 50 times higher than those of AGP, but AGP levels in plasma are increased by about 10 times under inflammatory disease. Therefore, the involvement of these 2 plasma proteins should be considered in more depth for understanding the pharmacokinetics of aripiprazole.
Collapse
Affiliation(s)
- Koji Nishi
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Keiki Sakurama
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Yoshihiro Kobashigawa
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi 5-1, Chuo-ku, Kumamoto 862-0082, Japan
| | - Hiroshi Morioka
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi 5-1, Chuo-ku, Kumamoto 862-0082, Japan
| | - Nagiko Udo
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Mai Hashimoto
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Shuhei Imoto
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan; DDS Research Institute, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan.
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan; DDS Research Institute, Sojo University, Ikeda 4-22-1, Nishi-ku, Kumamoto 860-0082, Japan.
| |
Collapse
|
10
|
Kono K, Fukuchi Y, Okawa H, Nunoya KI, Imawaka H, Watanabe H, Maruyama T. Unique Hydrolysis of an Ester-Type Prodrug of Levodopa in Human Plasma: Relay-Type Role Sharing between Alpha-1 Acid Glycoprotein and Human Serum Albumin. Mol Pharm 2019; 16:4131-4138. [DOI: 10.1021/acs.molpharmaceut.9b00435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Kenta Kono
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Ibaraki, Japan
| | - Yukina Fukuchi
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Ibaraki, Japan
| | - Hoshimi Okawa
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Ibaraki, Japan
| | - Ken-ichi Nunoya
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Ibaraki, Japan
| | - Haruo Imawaka
- Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Ibaraki, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
11
|
Amidon GE, Anderson BD, Balthasar JP, Bergstrom CAS, Huang SM, Kasting G, Kesisoglou F, Khinast JG, Mager DE, Roberts CJ, Yu L. Fifty-Eight Years and Counting: High-Impact Publishing in Computational Pharmaceutical Sciences and Mechanism-Based Modeling. J Pharm Sci 2018; 108:2-7. [PMID: 30423338 DOI: 10.1016/j.xphs.2018.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 11/19/2022]
Abstract
With this issue of the Journal of Pharmaceutical Sciences, we celebrate the nearly 6 decades of contributions to mechanistic-based modeling and computational pharmaceutical sciences. Along with its predecessor, The Journal of the American Pharmaceutical Association: Scientific Edition first published in 1911, JPharmSci has been a leader in the advancement of pharmaceutical sciences beginning with its inaugural edition in 1961. As one of the first scientific journals focusing on pharmaceutical sciences, JPharmSci has established a reputation for publishing high-quality research articles using computational methods and mechanism-based modeling. The journal's publication record is remarkable. With over 15,000 articles, 3000 notes, and more than 650 reviews from industry, academia, and regulatory agencies around the world, JPharmSci has truly been the leader in advancing pharmaceutical sciences.
Collapse
Affiliation(s)
| | | | - Joseph P Balthasar
- University at Buffalo, State University of New York, Buffalo, New York 14260
| | | | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | | | | | - Johannes G Khinast
- Institute for Process and Particle Engineering, Graz University of Technology, Graz, Austria
| | - Donald E Mager
- University at Buffalo, State University of New York, Buffalo, New York 14260
| | | | - Lian Yu
- University of Wisconsin, Madison, Wisconsin 53706
| |
Collapse
|
12
|
Concentration-dependent plasma protein binding: Expect the unexpected. Eur J Pharm Sci 2018; 122:341-346. [DOI: 10.1016/j.ejps.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 11/20/2022]
|
13
|
Schocken D, Stohlman J, Vicente J, Chan D, Patel D, Matta MK, Patel V, Brock M, Millard D, Ross J, Strauss DG, Blinova K. Comparative analysis of media effects on human induced pluripotent stem cell-derived cardiomyocytes in proarrhythmia risk assessment. J Pharmacol Toxicol Methods 2017; 90:39-47. [PMID: 29155283 DOI: 10.1016/j.vascn.2017.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/07/2017] [Accepted: 11/13/2017] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Cardiotoxicity assessment using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) forms a key component of the Comprehensive in Vitro Proarrhythmia Assay (CiPA). A potentially impactful factor on iPSC-CM testing is the presence of serum in the experimental media. Generally, serum-free media is used to most accurately reproduce "free" drug concentration. However, caution is needed; drug solubility and cardiomyocyte electrophysiology could be affected by media formulation, potentially impacting interpretation of drug-induced effects. METHODS Effects of 25 drugs on properties of spontaneous field potentials in iPSC-CMs were assayed using a high-throughput microelectrode array (MEA) in two media formulations: serum-containing and serum-free. Comparative analysis was conducted on rate-corrected field potential duration (FPDc) and prevalence of arrhythmic events. Further MEA experiments were conducted, varying percentages of serum as well as carbon substrate components. Comparative LC-MS/MS analysis was done on two compounds to evaluate drug concentrations. RESULTS In serum-free media, 9 drugs prolonged FPDc. In serum-containing, 11 drugs prolonged FPDc. Eighteen drugs induced arrhythmias, 8 of these induced arrhythmias at lower concentrations in serum-containing media. At the highest non-arrhythmic concentrations, 13 of 25 drugs exhibited significant differences in FPDc prolongation/shortening between the media. Increasing fractions of serum in media yielded higher FPDc measurements. LC-MS/MS analysis of moxifloxacin and quinidine showed higher concentrations in serum-containing media. DISCUSSION The present study highlights media formulation as an important consideration for cardiac safety testing with iPSC-CMs. Results described here suggest that media formulation influences both compound availability and baseline electrophysiological properties. Special attention should be paid to media for future iPSC-CM assays.
Collapse
Affiliation(s)
- Derek Schocken
- US Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biomedical Physics, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Jayna Stohlman
- US Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biomedical Physics, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Jose Vicente
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Division of Cardiovascular and Renal Products, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Dulciana Chan
- US Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biomedical Physics, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Dakshesh Patel
- US Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biomedical Physics, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Murali Krishna Matta
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Clinical Pharmacology, Division of Applied Regulatory Science, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Vikram Patel
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Clinical Pharmacology, Division of Applied Regulatory Science, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Mathew Brock
- Axion BioSystems, Inc., 1819 Peachtree Road NE, Suite 350, Atlanta, GA 30309, USA.
| | - Daniel Millard
- Axion BioSystems, Inc., 1819 Peachtree Road NE, Suite 350, Atlanta, GA 30309, USA.
| | - James Ross
- Axion BioSystems, Inc., 1819 Peachtree Road NE, Suite 350, Atlanta, GA 30309, USA.
| | - David G Strauss
- US Food and Drug Administration, Center for Drug Evaluation and Research, Office of Clinical Pharmacology, Division of Applied Regulatory Science, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Ksenia Blinova
- US Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Biomedical Physics, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| |
Collapse
|
14
|
Mora-Gutierrez A, Attaie R, Núñez de González MT, Jung Y, Woldesenbet S, Marquez SA. Complexes of lutein with bovine and caprine caseins and their impact on lutein chemical stability in emulsion systems: Effect of arabinogalactan. J Dairy Sci 2017; 101:18-27. [PMID: 29103708 DOI: 10.3168/jds.2017-13105] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/06/2017] [Indexed: 11/19/2022]
Abstract
Lutein is an important xanthophyll carotenoid with many benefits to human health. Factors affecting the application of lutein as a functional ingredient in low-fat dairy-like beverages (pH 6.0-7.0) are not well understood. The interactions of bovine and caprine caseins with hydrophobic lutein were studied using UV/visible spectroscopy as well as fluorescence. Our studies confirmed that the aqueous solubility of lutein is improved after binding with bovine and caprine caseins. The rates of lutein solubilization by the binding to bovine and caprine caseins were as follows: caprine αS1-II-casein 34%, caprine αS1-I-casein 10%, and bovine casein 7% at 100 μM lutein. Fluorescence of the protein was quenched on binding supporting complex formation. The fluorescence experiments showed that the binding involves tryptophan residues and some nonspecific interactions. Scatchard plots of lutein binding to the caseins demonstrated competitive binding between the caseins and their sites of interaction with lutein. Competition experiments suggest that caprine αS1-II casein will bind a larger number of lutein molecules with higher affinity than other caseins. The chemical stability of lutein was largely dependent on casein type and significant increases occurred in the chemical stability of lutein with the following pattern: caprine αS1-II-casein > caprine αS1-I-casein > bovine casein. Addition of arabinogalactan to lutein-enriched emulsions increases the chemical stability of lutein-casein complexes during storage under accelerated photo-oxidation conditions at 25°C. Therefore, caprine αS1-II-casein alone and in combination with arabinogalactan can have important applications in the beverage industry as carrier of this xanthophyll carotenoid (lutein).
Collapse
Affiliation(s)
- A Mora-Gutierrez
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446.
| | - R Attaie
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446
| | - M T Núñez de González
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446
| | - Y Jung
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446
| | - S Woldesenbet
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446
| | - S A Marquez
- Cooperative Agricultural Research Center, Prairie View A&M University, Prairie View, TX 77446
| |
Collapse
|
15
|
Characterization of the interaction of glycyrrhizin and glycyrrhetinic acid with bovine serum albumin by spectrophotometric-gradient flow injection titration technique and molecular modeling simulations. Int J Biol Macromol 2017; 102:92-103. [PMID: 28377238 DOI: 10.1016/j.ijbiomac.2017.02.106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 01/19/2023]
Abstract
In this research, the interactions of glycyrrhizin (GL) and glycyrrhetinic acid (GA) with bovine serum albumin (BSA) have been investigated by the novel method of spectrophotometric- gradient flow injection titration technique. The hard-modeling multivariate approach to binding was used for calculation of binding constants and estimation of concentration-spectral profiles of equilibrium species. The stoichiometric ratio of binding was estimated using eigenvalue analysis. Results showed that GL and GA bind BSA with overall binding constants of KGL-BSA=3.85 (±0.09)×104Lmol-1, KGA-BSA=3.08 (±0.08)×104Lmol-1. Ligand-BSA complexes were further analyzed by combined docking and molecular dynamics (MD) simulations. Docking simulations were performed to obtain a first guess on the binding structure of the GL/GA-BSA complex, and subsequently analyzed by 20 ns MD simulations in order to evaluate interactions of GL/GA with BSA in detail. Results of MD simulations indicated that GL-BSA complex forms mainly on the basis of hydrogen bonds, while, GA-BSA complex forms on the basis of hydrophobic interactions. Also, water molecules can bridge between the ligand and protein by hydrogen bonds, which are stable during the entire simulation and play an important role in stabilization of the GL/GA-BSA complexes.
Collapse
|
16
|
Manouchehri F, Izadmanesh Y, Aghaee E, Ghasemi JB. Experimental, computational and chemometrics studies of BSA-vitamin B6 interaction by UV–Vis, FT-IR, fluorescence spectroscopy, molecular dynamics simulation and hard-soft modeling methods. Bioorg Chem 2016; 68:124-36. [DOI: 10.1016/j.bioorg.2016.07.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/13/2022]
|
17
|
Yan J, Wu D, Sun P, Ma X, Wang L, Li S, Xu K, Li H. Binding mechanism of the tyrosine-kinase inhibitor nilotinib to human serum albumin determined by 1 H STD NMR, 19 F NMR, and molecular modeling. J Pharm Biomed Anal 2016; 124:1-9. [DOI: 10.1016/j.jpba.2016.02.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/15/2016] [Accepted: 02/17/2016] [Indexed: 11/28/2022]
|
18
|
Morris TT, Ruan Y, Lewis VA, Narendran A, Gailer J. Fortification of blood plasma from cancer patients with human serum albumin decreases the concentration of cisplatin-derived toxic hydrolysis products in vitro. Metallomics 2015; 6:2034-41. [PMID: 25255207 DOI: 10.1039/c4mt00220b] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While cisplatin (CP) is still one of the world's bestselling anticancer drugs, its intravenous administration is inherently associated with severe, dose limiting toxic side-effects. Although the molecular basis of the latter are not well understood, biochemical transformations of CP in blood and the interaction of the generated platinum species with plasma proteins likely play a critical role since these processes will ultimately determine which platinum-species reach the intended tumor cells as well as non-target cells. Compared to healthy subjects, cancer patients often have decreased plasma human serum albumin (HSA) concentrations. Little, however, is known about how the plasma HSA concentration will affect the metabolism of CP. To gain insight, we obtained blood plasma from healthy adults (n = 20, 42 ± 4 g HSA per L) and pediatric cancer patients (n = 11, 26 ± 7 g HSA per L). After the incubation of plasma at 37 °C, a pharmacologically relevant dose of CP was added and the Pt-distribution therein was determined by size-exclusion chromatography coupled on-line to an inductively coupled plasma atomic emission spectrometer. At the 2 h time point, a 5.9% increase of toxic CP-derived hydrolysis products was detected in pediatric cancer patient plasma, while 9.8% less platinum was protein bound compared to plasma from healthy controls. These in vitro results suggest that the elevated concentration of highly reactive free CP-derived hydrolysis products in plasma may cause the toxic side-effects in cancer patients. More importantly, the deliberate increase of the plasma HSA concentration in cancer patients prior to CP treatment would represent a simple strategy to possibly alleviate the fraction of patients that suffer from drug induced toxic side-effects.
Collapse
Affiliation(s)
- Thomas T Morris
- Department of Chemistry, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | | | | | | | | |
Collapse
|
19
|
Garzón A, Bravo I, Carrión-Jiménez MR, Rubio-Moraga Á, Albaladejo J. Spectroscopic study on binding of gentisic acid to bovine serum albumin. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 150:26-33. [PMID: 26010705 DOI: 10.1016/j.saa.2015.05.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 06/04/2023]
Abstract
The interaction of (gentisic acid) GA with (bovine serum albumin) BSA has been studied by different spectroscopic techniques. GA is a monoanionic specie at the working pH of 7.4, it was determined by combining UV-Vis absorption spectroscopy and theoretical calculations. A set of fluorescence quenching experiments at different temperatures was carried out employing the native fluorescence of BSA. A Stern-Volmer constant (KSV) of (2.07±0.12)×10(4) mol(-1) L and a binding constant (Ka) of (8.47±4.39)×10(3) were determined at 310 K. The static quenching caused by the BSA-GA complex formation seems to play a significant role in the overall quenching process. A single binding site on BSA for GA was observed. ΔH=-55.6±0.2 kJ mol(-1) and ΔS=-104.3±0.6 J mol(-1) K(-1) were determined in a set of experiments on the dependence of Ka with the temperature. The binding process is, therefore, spontaneous and enthalpy-driven. Van der Waals forces and hydrogen bonds could also play the major role in the binding mode. The secondary structure changes of BSA in the absence and presence of GA were studied by FTIR and UV-Vis absorption spectroscopy.
Collapse
Affiliation(s)
- Andrés Garzón
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Paseo de los estudiantes, s/n, 02071 Albacete, Spain.
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Paseo de los estudiantes, s/n, 02071 Albacete, Spain
| | - M Rosario Carrión-Jiménez
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Paseo de los estudiantes, s/n, 02071 Albacete, Spain
| | - Ángela Rubio-Moraga
- Instituto Botánico, Facultad de Farmacia, Universidad de Castilla-La Mancha, Paseo de los estudiantes, s/n, 02071 Albacete, Spain
| | - José Albaladejo
- Departamento de Química Física, Facultad de Ciencias Químicas, Universidad de Castilla-La Mancha, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| |
Collapse
|
20
|
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]
|
21
|
Interactive association of drugs binding to human serum albumin. Int J Mol Sci 2014; 15:3580-95. [PMID: 24583848 PMCID: PMC3975355 DOI: 10.3390/ijms15033580] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/17/2014] [Accepted: 02/18/2014] [Indexed: 02/06/2023] Open
Abstract
Human serum albumin (HSA) is an abundant plasma protein, which attracts great interest in the pharmaceutical industry since it can bind a remarkable variety of drugs impacting their delivery and efficacy and ultimately altering the drug’s pharmacokinetic and pharmacodynamic properties. Additionally, HSA is widely used in clinical settings as a drug delivery system due to its potential for improving targeting while decreasing the side effects of drugs. It is thus of great importance from the viewpoint of pharmaceutical sciences to clarify the structure, function, and properties of HSA–drug complexes. This review will succinctly outline the properties of binding site of drugs in IIA subdomain within the structure of HSA. We will also give an overview on the binding characterization of interactive association of drugs to human serum albumin that may potentially lead to significant clinical applications.
Collapse
|
22
|
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
|
23
|
Nishi K, Kobayashi M, Nishii R, Shikano N, Takamura N, Kuga N, Yamasaki K, Nagamachi S, Tamura S, Otagiri M, Kawai K. Pharmacokinetic Alteration of 99mTc-MAG3 using Serum Protein Binding Displacement Method. Nucl Med Biol 2013; 40:366-70. [DOI: 10.1016/j.nucmedbio.2012.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 11/26/2012] [Accepted: 12/01/2012] [Indexed: 10/27/2022]
|
24
|
Depot-Protection Against γ-Irradiation in Mice by Parenteral Glutathione-Polyvinylpyrrolidone Coprecipitates and Cellular Drug Uptake. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
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
|
26
|
Roch‐Ramel F, Besseghir K, Murer H. Renal Excretion and Tubular Transport of Organic Anions and Cations. Compr Physiol 2011. [DOI: 10.1002/cphy.cp080248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Ghosh A, Nandy A. Graphical representation and mathematical characterization of protein sequences and applications to viral proteins. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2011; 83:1-42. [PMID: 21570664 PMCID: PMC7150266 DOI: 10.1016/b978-0-12-381262-9.00001-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Graphical representation and numerical characterization (GRANCH) of nucleotide and protein sequences is a new field that is showing a lot of promise in analysis of such sequences. While formulation and applications of GRANCH techniques for DNA/RNA sequences started just over a decade ago, analyses of protein sequences by these techniques are of more recent origin. The emphasis is still on developing the underlying technique, but significant results have been achieved in using these methods for protein phylogeny, mass spectral data of proteins and protein serum profiles in parasites, toxicoproteomics, determination of different indices for use in QSAR studies, among others. We briefly mention these in this chapter, with some details on protein phylogeny and viral diseases. In particular, we cover a systematic method developed in GRANCH to determine conserved surface exposed peptide segments in selected viral proteins that can be used for drug and vaccine targeting. The new GRANCH techniques and applications for DNAs and proteins are covered briefly to provide an overview to this nascent field.
Collapse
Affiliation(s)
- Ambarnil Ghosh
- Physics Department, Jadavpur University, Jadavpur, Kolkata, India
| | | |
Collapse
|
28
|
Abstract
After being distributed in the circulating blood, drugs bind to serum proteins varying degrees. In general, such binding is reversible, and a dynamic equilibrium exists between the bound and unbound molecular species. It is believed that unless there is a specific transport system (e.g. receptor-mediated endocytosis, protein-mediated transport), only unbound drugs are able to penetrate through biomembranes, are distributed to tissues, and undergo metabolism and glomerular filtration. It is also believed that only unbound molecules present in target tissues can exert their pharmacological effects, and that the concentration of unbound molecules in tissues is in proportion to the drug serum concentration. Therefore, drug-serum protein binding is critically involved in the manifestation of the pharmacological effects of a drug as well as its pharmacokinetics. Among serum proteins, human serum albumin (HSA) and alpha(1)-acid glycoprotein (AGP) play important roles in protein binding for many drugs, which is of key importance to drug distribution in the body. In addition, they are widely used in clinical settings as blood preparations and drug delivery system carriers. It is thus of great importance from the viewpoint of pharmaceutical science to clarify the structure, function, and pharmaceutical properties of HSA and AGP. Accordingly, since starting my laboratory, the focus of my research has involved molecular pharmaceutical studies on the interactions of drugs and HSA and AGP for the purpose of applying these findings to clinical fields, such as drug treatment, diagnosis and drug discovery. In this review, the molecular properties of HSA and AGP will be briefly outlined. The static and dynamic topology of drug binding sites on these proteins, investigated by various spectroscopic techniques, X-ray crystallography, quantitative structure-activity relationships, molecular modeling, photo affinity labeling, site-directed mutagenesis etc., changes in the serum protein binding of drugs in pathological conditions, such as liver and kidney failure and various inflammation diseases and factors contributing to the changes will then be summarized. Finally, cases in which protein binding displacement can be applied to medical fields will also be introduced.
Collapse
Affiliation(s)
- Masaki Otagiri
- Faculty of Medical and Pharmaceutical Sciences, Kumamoto University, Japan.
| |
Collapse
|
29
|
Andreasen F. Protein binding of drugs in plasma from patients with acute renal failure. ACTA PHARMACOLOGICA ET TOXICOLOGICA 2009; 32:417-29. [PMID: 4800749 DOI: 10.1111/j.1600-0773.1973.tb01488.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
30
|
Agren A, Elofsson R, Nilsson SO. Some physico-chemical factors influencing the binding of sulfonamides to human albumin in vitro. ACTA PHARMACOLOGICA ET TOXICOLOGICA 2009; 29 Suppl 3:48-56. [PMID: 5316410 DOI: 10.1111/j.1600-0773.1971.tb03284.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
31
|
Protein binding study of catechin hydrate and genistein by high-performance frontal analysis. KOREAN J CHEM ENG 2008. [DOI: 10.1007/s11814-008-0242-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
32
|
El-Nimr AE. Studies on Sulfaethidole - Lysozyme Interactions by Fluorescence Quenching. Drug Dev Ind Pharm 2008. [DOI: 10.3109/03639048409040787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
Chignell CF, Weber WW. Application of Physicochemical and Analytic Techniques to the Study of Drug Interactions with Biological Systems. ACTA ACUST UNITED AC 2008. [DOI: 10.3109/10408447209103466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
|
35
|
|
36
|
Østergaard J, Larsen C. Bioreversible derivatives of phenol. 2. Reactivity of carbonate esters with fatty acid-like structures towards hydrolysis in aqueous solutions. Molecules 2007; 12:2396-412. [PMID: 17978765 DOI: 10.3390/12102396] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 10/29/2007] [Accepted: 10/29/2007] [Indexed: 01/26/2023] Open
Abstract
A series of model phenol carbonate ester prodrugs encompassing derivatives with fatty acid-like structures were synthesized and their stability as a function of pH (range 0.4 - 12.5) at 37 degrees C in aqueous buffer solutions investigated. The hydrolysis rates in aqueous solutions differed widely, depending on the selected pro-moieties (alkyl and aryl substituents). The observed reactivity differences could be rationalized by the inductive and steric properties of the substituent groups when taking into account that the mechanism of hydrolysis may change when the type of pro-moiety is altered, e.g. n-alkyl vs. t-butyl. Hydrolysis of the phenolic carbonate ester 2-(phenoxycarbonyloxy)-acetic acid was increased due to intramolecular catalysis, as compared to the derivatives synthesized from omega-hydroxy carboxylic acids with longer alkyl chains. The carbonate esters appear to be less reactive towards specific acid and base catalyzed hydrolysis than phenyl acetate. The results underline that it is unrealistic to expect that phenolic carbonate ester prodrugs can be utilized in ready to use aqueous formulations. The stability of the carbonate ester derivatives with fatty acid-like structures, expected to interact with the plasma protein human serum albumin, proved sufficient for further in vitro and in vivo evaluation of the potential of utilizing HSA binding in combination with the prodrug approach for optimization of drug pharmacokinetics.
Collapse
Affiliation(s)
- Jesper Østergaard
- Department of Pharmaceutics and Analytical Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen O, Denmark.
| | | |
Collapse
|
37
|
Østergaard J, Larsen C. Bioreversible derivatives of phenol. 1. The role of human serum albumin as related to the stability and binding properties of carbonate esters with fatty acid-like structures in aqueous solution and biological media. Molecules 2007; 12:2380-95. [PMID: 17978764 PMCID: PMC6149159 DOI: 10.3390/12102380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 10/29/2007] [Accepted: 10/29/2007] [Indexed: 11/17/2022] Open
Abstract
With the overall objective of assessing the potential of utilizing plasma protein binding interactions in combination with the prodrug approach for improving the pharmacokinetics of drug substances, a series of model carbonate ester prodrugs of phenol, encompassing derivatives with fatty acid-like structures, were characterized in vitro. Stability of the derivatives was studied in aqueous solution, human serum albumin solution, human plasma, and rat liver homogenate at 37 degrees C. Stability of the derivatives in aqueous solution varied widely, with half-lives ranging from 31 to 1.7 x 10(4) min at pH 7.4 and 37 degrees C. The carbonate esters were subject to catalysis by plasma esterases except for the t-butyl and acetic acid derivatives, which were stabilized in both human plasma and human serum albumin solutions relative to buffer. In most cases, however, hydrolysis was accelerated in the presence of human serum albumin indicating that the derivatives interacted with the protein, a finding which was confirmed using the p-nitrophenyl acetate kinetic assay. Different human serum albumin binding properties of the phenol model prodrugs with fatty acid-like structure and neutral carbonate esters were observed. In the context of utilizing plasma protein binding in combination with the prodrug approach for optimizing drug pharmacokinetics, the esterase-like properties of human serum albumin towards the carbonate esters potentially allowing the protein to act as a catalyst of parent compound regenerations is interesting.
Collapse
Affiliation(s)
- Jesper Østergaard
- Department of Pharmaceutics and Analytical Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen O, Denmark; E-mail:
| | - Claus Larsen
- Department of Pharmaceutics and Analytical Chemistry, Faculty of Pharmaceutical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen O, Denmark; E-mail:
| |
Collapse
|
38
|
Platé NA, Valuev LI. Polymeric biospecific adsorbents for binding some blood components. ACTA ACUST UNITED AC 2007. [DOI: 10.1002/polc.5070660116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
39
|
|
40
|
Eder AR, Chen JS, Arriaga EA. Separation of doxorubicin and doxorubicinol by cyclodextrin-modified micellar electrokinetic capillary chromatography. Electrophoresis 2006; 27:3263-70. [PMID: 16915573 DOI: 10.1002/elps.200600025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Doxorubicinol (DOXol) is a human metabolite of the chemotherapy agent doxorubicin (DOX), and is associated with dose-dependent cardiotoxicity and decreased drug efficacy. Due to the structural similarities and equal molecular charges of DOXol and DOX, their electrophoretic separation is commonly ineffective. A method for separating and detecting DOX and DOXol, as well as two DOXol enantiomers, was established using cyclodextrin-modified micellar electrokinetic capillary chromatography with laser-induced fluorescence detection. Differential DOXol production was detected in a DOX-sensitive and resistant pair of cell lines, with a 0.08 +/- 0.01 fmol limit of detection.
Collapse
Affiliation(s)
- Angela R Eder
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
41
|
Otagiri M. A molecular functional study on the interactions of drugs with plasma proteins. Drug Metab Pharmacokinet 2006; 20:309-23. [PMID: 16272748 DOI: 10.2133/dmpk.20.309] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The binding of drugs to plasma proteins, such as albumin and alpha1-acid glycoprotein (AGP) is a major determinant in the disposition of drugs. A topology analysis of drug binding sites on HSA and AGP was determined using various methods, including spectroscopy, QSAR, photoaffinity labeling and site directed mutagenesis. Recombinant albumin was found to be useful for rapidly identifying drug binding sites. The binding sites on AGP are not completely separated but are partially overlapped, and Trp, Tyr, Lys and His residues in the drug binding pockets play important roles in this process. Drug displacement is somewhat complex, due to the involvement of multiple effects. The reduced binding in uremic patients may be explained by a mechanism that involves a combination of direct displacement by free fatty acids as well as cascade effects of free fatty acids and unbound uremic toxins for significant inhibition in serum binding. Albumin-containing dialysate is useful for the extracorporeal removal of endogenous toxins and in the treatment of drug overdoses. Oxidized albumin is a useful biomarker for the quantitative and qualitative evaluation of oxidative stress. Interestingly, AGP undergoes a structural transition to a unique structure that differs from the native and denatured states, when it interacts with membranes.
Collapse
Affiliation(s)
- Masaki Otagiri
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi, Japan.
| |
Collapse
|
42
|
|
43
|
Takamura N, Maruyama T, Chosa E, Kawai K, Tsutsumi Y, Uryu Y, Yamasaki K, Deguchi T, Otagiri M. BUCOLOME, A POTENT BINDING INHIBITOR FOR FUROSEMIDE, ALTERS THE PHARMACOKINETICS AND DIURETIC EFFECT OF FUROSEMIDE: POTENTIAL FOR USE OF BUCOLOME TO RESTORE DIURETIC RESPONSE IN NEPHROTIC SYNDROME. Drug Metab Dispos 2005; 33:596-602. [PMID: 15640375 DOI: 10.1124/dmd.104.002782] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To determine whether bucolome (5-n-butyl-1-cyclohexyl-2,4,6-trioxoperhydropyrimidine), a nonsteroidal anti-inflammatory agent, can reverse diuretic resistance of furosemide in patients with nephrotic syndrome, we examined the inhibitory effect of bucolome on the protein binding of furosemide in serum and urine. Bucolome significantly inhibited the protein binding of furosemide not only in serum but also in urine of preparation albumin (UPA), which mimics urinary albumin concentration in patients with nephrotic syndrome by ultrafiltration method. The binding percentage of furosemide to albumin was approximately 70% in UPA. With coadministration of bucolome to healthy volunteers, renal clearance of furosemide was increased, reflecting the increase of the free fraction of furosemide in serum. Furthermore, coadministration of bucolome caused a significant increase of urine volume and sodium concentration in urine. Even at higher urine levels of furosemide, the inhibitory effect of bucolome on the protein binding of furosemide in UPA remains constant, and changes in pH at weakly acidic pH levels (pH 5.5-6.5) did not alter the inhibitory effect of bucolome. Interestingly, coadministration of bucolome with furosemide in doxorubicin (Adriamycin)-induced nephrotic syndrome model rats alleviated the diuretic resistance. These results suggest that bucolome has a potent inhibitory effect on the protein binding of furosemide in the urine and can partially restore the diuretic response of furosemide in patients with nephrotic syndrome by increasing the free fraction of furosemide at the site of action.
Collapse
Affiliation(s)
- Norito Takamura
- Department of Pharmacy, Miyazaki Medical College Hospital, Miyazaki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kuroda Y, Shibukawa A, Nakagawa T. Drug Binding Analysis of Human α 1-Acid Glycoprotein Using Capillary Electrophoresis. YAKUGAKU ZASSHI 2003; 123:781-8. [PMID: 14513769 DOI: 10.1248/yakushi.123.781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Drug-plasma protein binding analysis is indispensable for drug development and clinical use. However, conventional methods for binding analyses were not suitable for small amounts of proteins because of large sample requirements. On the other hand, high-performance frontal analysis/capillary electrophoresis (HPFA/CE) consumes very small sample volumes, and is useful for ligand-binding study of small amounts of proteins. In this study, HPFA/CE was used in a drug-binding study of alpha 1-acid glycoprotein (AGP) subtypes in which plasma concentrations change dynamically to elucidate the effects of structural variation on drug binding. Binding study on desialyrated AGP revealed that (S)-enantiomer selectivity in propranolol-AGP binding was caused by sialic acid residues, while neither sialic acid nor galactose caused the enantioselectivity of verapamil binding to AGP. Biantennary glycans slightly suppressed disopyramide binding to AGP, whereas the glycans did not have any influence on propranolol and verapamil binding. Disopyramide and verapamil were selectively bound to the A variant rather than the F1S variant. The A variant showed larger enantioselective binding to disopyramide, but not to verapamil.
Collapse
Affiliation(s)
- Yukihiro Kuroda
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | | | | |
Collapse
|
45
|
Kuroda Y, Matsumoto S, Shibukawa A, Nakagawa T. Capillary electrophoretic study on pH dependence of enantioselective disopyramide binding to genetic variants of human alpha1-acid glycoprotein. Analyst 2003; 128:1023-7. [PMID: 12964601 DOI: 10.1039/b212850k] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A high-performance frontal analysis-capillary electrophoresis (HPFA-CE) method was applied to investigate the effect of pH on the drug binding properties of genetic variants of human alpha1-acid glycoprotein (AGP), A variant and a mixture of F1S variants. The unbound concentrations of a model basic drug, disopyramide (DP), in A variant solutions and in F1S variant solutions were measured by HPFA-CE to evaluate binding constants at pH 4.0, 5.0, 6.0 and 7.4. The binding between DP and A variant was gradually weakened by acidification of background buffer (from pH 7.4 to 4.0), while the binding between DP and FIS variants decreased at first (from pH 7.4 to 6.0), and then gained (from pH 6.0 to 4.0). Consequently, DP was more strongly bound to A variant than to FIS variants at pH 7.4, while at pH 4.0 DP was more strongly bound to F1S variants. At any pH (S)-DP was bound more strongly than (R)-DP, and the enantioselectivity of A variant was significantly higher than that of F1S variants. Electrophoretic mobilities of the AGP genetic variants decreased along with a decrease in pH. Fluorescent emission of these genetic variants indicated a distinct conformational change between pH 5.0 and 4.0. However, there was no significant difference in the electrophoretic mobility and the fluorescent emission spectrum between these variants at any pH. On the other hand, circular dichroism analyses revealed that beta-sheet content in FIS variants diminished as pH decreased, while that in A variant increased. These results suggest that the conformational change induced by acidification of background buffer differs between these genetic variants, and this causes the difference in DP bindability.
Collapse
Affiliation(s)
- Yukihiro Kuroda
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.
| | | | | | | |
Collapse
|
46
|
Kuroda Y, Watanabe Y, Shibukawa A, Nakagawa T. Role of phospholipids in drug-LDL bindings as studied by high-performance frontal analysis/capillary electrophoresis. J Pharm Biomed Anal 2003; 30:1869-77. [PMID: 12485729 DOI: 10.1016/s0731-7085(02)00530-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The binding study between basic drugs ((S)-verapamil (VER) and (S)-propranolol (PRO)) and phospholipid liposomes was performed by using high-performance frontal analysis/capillary electrophoresis (HPFA/CE) in order to investigate the effect of oxidative modification of low-density lipoprotein (LDL) upon drug-binding affinity from molecule-based viewpoint. 1-Palmitoyl-2-oleoyl-phosphatidylcholine (POPC, 16:0, 18:1), 1-palmitoyl-2-linoleoyl-phosphatidylcholine (PLPC, 16:0, 18:2), dilauloyl-phosphatidylcholine (DLaPC, 12:0, 12:0), 1-palmitoyl-2-oleoyl-phosphatidyl-glycerol (POPG, 16:0, 18:1), and 1-palmitoyl-sn-glycero-3-phosphocholine (monoPPC, 16:0) were used to prepare the model liposomes. At physiological pH (pH 7.4), the model liposome prepared from POPG+POPC had negative net charges, while the total net charge of the other model liposomes (POPC liposome, PLPC liposome, DLaPC liposome, and monoPPC+POPC liposome) was zero. The drug and the model liposome mixed solutions were subjected to HPFA/CE, and the total binding affinities (nK) were calculated. The nK values of VER and PRO to POPG+POPC liposome were more than six and 10 times higher than those of other liposomes, respectively. On the other hand, the nK values of the model drugs to POPC liposome, PLPC liposome, DLaPC liposome and monoPPC+POPC liposome showed small differences less than twice. These results indicate that the electrostatic interaction plays an important effect on drug-liposome binding, and suggest that the increase in the negative charge of LDL phospholipids gives more significant effect on the drug-binding affinity of the basic drugs than the acyl-chain structure.
Collapse
Affiliation(s)
- Yukihiro Kuroda
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | |
Collapse
|
47
|
Li F, Qiao M, Guo X. Specific determination of unbound oxacillin in protein solution with cefoperazone by high-performance frontal analysis with chemiluminescence detection. Biomed Chromatogr 2003; 17:53-7. [PMID: 12583007 DOI: 10.1002/bmc.213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Unbound oxacillin concentrations in human serum albumin (HSA) solutions in the presence or absence of cefoperazone were determined using high-performance frontal analysis coupled with chemiluminescence detection (HPFA-CL). The HPFA was performed on an ISRP column with 67 mM potassium phosphate buffer of pH 7.4 and ionic strength of 0.17 as the mobile phase. The luminol-H(2)O(2)-Co(2+) system was employed in the chemiluminescence detection. The detection was highly specific for oxacillin in the presence of cefoperazone. Although both drugs in HSA solutions co-eluted in the same region in HPFA, cefoperazone did not interfere with the determination of unbound concentration of oxacillin. In the solution of 100 microM HSA and 11.33 micro M oxacillin the bound percentage of oxacillin to HSA was estimated as 80.5%. Addition of 30.98 micro M cefoperazone into the HSA-equilibrated solution produced little effect on the protein binding of oxacillin. In the presence of 154.9 micro M cefoperazone, however, the bound percentage of oxacillin was significantly reduced. This specific method could be applied to the investigation of drug-drug interaction in protein binding.
Collapse
Affiliation(s)
- Famei Li
- Department of Analytical Chemistry, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, People's Republic of China.
| | | | | |
Collapse
|
48
|
Qiao M, Guo X, Li F. Chemiluminescence detection coupled to high-performance frontal analysis for the determination of unbound concentrations of drugs in protein binding equilibrium. J Chromatogr A 2002; 952:131-8. [PMID: 12064525 DOI: 10.1016/s0021-9673(02)00086-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
High-performance frontal analysis coupled with chemiluminescence detection (HPFA-CL) was developed for the determination of unbound oxacillin concentration in human serum albumin solution. The HPFA system consisted of an ISRP column and a mobile phase of 67 mM potassium phosphate buffer of pH 7.4 and ionic strength of 0.17. The luminol-H2O2-Co2+ system was used in the chemiluminescence detection. An enhancement of luminol chemiluminescence by oxacillin was investigated and employed for determining the concentration of oxacillin in the HPFA eluate. Sample solutions were directly injected onto the column; the drug was eluted as a zonal peak with a plateau region. The unbound drug concentrations were determined by using the height of the plateau. The results agreed with those obtained with conventional ultrafiltration-HPLC method. Good reproducibility was confirmed by the within run and between run RSD < or = 7.4%. HPFA-CL provided a selective method for determination of unbound drug concentration in protein binding equilibrium.
Collapse
Affiliation(s)
- Mingxi Qiao
- Department of Analytical Chemistry, Shenyang Pharmaceutical University, China
| | | | | |
Collapse
|
49
|
Shibukaw A, Yoshikawa Y, Kimura T, Kuroda Y, Nakagawa T, Wainer IW. Binding study of desethyloxybutynin using high-performance frontal analysis method. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 768:189-97. [PMID: 11939552 DOI: 10.1016/s0378-4347(01)00499-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Plasma protein binding of N-desethyloxybytynin (DEOXY), a major active metabolite of oxybutynin (OXY), was investigated quantitatively and enantioselectively using high-performance frontal analysis (HPFA). An on-line HPLC system which consists of HPFA column, extraction column and analytical column was developed to determine the unbound concentrations of DEOXY enantiomers in human plasma, in human serum albumin (HSA) solutions, and in human alpha1-acid glycoprotein (AGP) solutions. DEOXY is bound in human plasma strongly and enantioselectively. The unbound drug fraction in human plasma samples containing 5 microM (R)- or (S)-DEOXY was 1.19 +/- 0.001 and 2.33 +/- 0.044%, respectively. AGP plays the dominant role in this strong and enantioselective plasma protein binding of DEOXY. The total binding affinity (nK) of (R)-DEOXY and (S)-DEOXY to AGP was 2.97 x 10(7) and 1.31 x 10(7) M(-1), respectively, while the nK values of (R)-DEOXY and (S)-DEOXY to HSA were 7.77 x 10(3) and 8.44 x 10(3) M(-1), respectively. While the nK value of (S)-DEOXY is weaker than that of (S)-OXY (1.53 x 10(7) M(-1)), the nK value of (R)-DEOXY is 4.33 times stronger than that of (R)-OXY (6.86 x I0(6) M(-1)). This suggests that the elimination of an ethyl group weakens the binding affinity of the (S)-isomer because of the decrease in hydrophobicity, while the binding affinity of the (R)-isomer is enhanced by the decrease in steric hindrance. The total binding affinity of DEOXY to HSA is much lower than that of DEOXY-AGP binding as well as OXY-HSA binding (2.64 x 10(4) and 2.19 x 10(4) M(-1) for (R)-OXY and (S)-OXY, respectively). The study on competitive binding between OXY and DEOXY indicated that DEOXY enantiomers and OXY enantiomers are all bound competitively at the same binding site of AGP molecule.
Collapse
Affiliation(s)
- Akimasa Shibukaw
- Graduate School of Pharmaceutical Sciences, Kyoto University, Japan.
| | | | | | | | | | | |
Collapse
|
50
|
Shibukawa A, Ishizawa N, Kimura T, Sakamoto Y, Ogita K, Matsuo Y, Kuroda Y, Matayatsu C, Nakagawa T, Wainer IW. Plasma protein binding study of oxybutynin by high-performance frontal analysis. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 768:177-88. [PMID: 11939551 DOI: 10.1016/s0378-4347(01)00497-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Plasma protein binding of oxybutynin (OXY) was investigated quantitatively and enantioselectively using high-performance frontal analysis (HPFA). An on-line HPLC system which consists of HPFA column, extraction column and analytical column was developed to determine the unbound concentrations of OXY enantiomers in human plasma, in human serum albumin (HSA) solutions, and in human alpha1-acid glycoprotein (AGP) solutions. OXY is bound in human plasma strongly and enantioselectively. The bound drug fraction in human plasma containing 2-10 microM (R)- or (S)-OXY was higher than 99%, and the unbound fraction of (R)-OXY was 1.56 times higher than that of (S)-isomer. AGP plays the dominant role in this strong and enantioselective plasma protein binding. The total binding affinities (nK) of (R)- and (S)-OXY to AGP were 6.86 x 10(6) and 1.53 x 10(7) M(-1), respectively, while the nK values of (R)- and (S)-OXY to HSA were 2.64 x 10(4) and 2.19 x 10(-4) M(-1), respectively. The binding affinity of OXY to AGP is much higher than that to HSA, and shows high enantioselectivity (SIR ratio of nK values is 2.2). It was found that both enantiomers are bound competitively at the same binding site on an AGP molecule. The binding property between OXY and low density lipoprotein (LDL) was investigated by using the frontal analysis method incorporated in high-performance capillary electrophoresis (HPCE/FA). It was found the binding is non-saturable and non-enantioselective.
Collapse
Affiliation(s)
- Akimasa Shibukawa
- Graduate School of Pharmaceutical Sciences, Kyoto University, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|