1
|
Hameed P S, Kotakonda H, Sharma S, Nandishaiah R, Katagihallimath N, Rao R, Sadler C, Slater I, Morton M, Chandrasekaran A, Griffen E, Pillai D, Reddy S, Bharatham N, Venkatesan S, Jonnalagadda V, Jayaraman R, Nanjundappa M, Sharma M, Raveendran S, Rajagopal S, Tumma H, Watters A, Becker H, Lindley J, Flamm R, Huband M, Sahm D, Hackel M, Mathur T, Kolamunnage-Dona R, Unsworth J, Mcentee L, Farrington N, Manickam D, Chandrashekara N, Jayachandiran S, Reddy H, Shanker S, Richard V, Thomas T, Nagaraj S, Datta S, Sambandamurthy V, Ramachandran V, Clay R, Tomayko J, Das S, V B. BWC0977, a broad-spectrum antibacterial clinical candidate to treat multidrug resistant infections. Nat Commun 2024; 15:8202. [PMID: 39294149 PMCID: PMC11410943 DOI: 10.1038/s41467-024-52557-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/12/2024] [Indexed: 09/20/2024] Open
Abstract
The global crisis of antimicrobial resistance (AMR) necessitates the development of broad-spectrum antibacterial drugs effective against multi-drug resistant (MDR) pathogens. BWC0977, a Novel Bacterial Topoisomerase Inhibitor (NBTI) selectively inhibits bacterial DNA replication via inhibition of DNA gyrase and topoisomerase IV. BWC0977 exhibited a minimum inhibitory concentration (MIC90) of 0.03-2 µg/mL against a global panel of MDR Gram-negative bacteria including Enterobacterales and non-fermenters, Gram-positive bacteria, anaerobes and biothreat pathogens. BWC0977 retains activity against isolates resistant to fluoroquinolones (FQs), carbapenems and colistin and demonstrates efficacy against multiple pathogens in two rodent species with significantly higher drug levels in the epithelial lining fluid of infected lungs. In healthy volunteers, single-ascending doses of BWC0977 administered intravenously ( https://clinicaltrials.gov/study/NCT05088421 ) was found to be safe, well tolerated (primary endpoint) and achieved dose-proportional exposures (secondary endpoint) consistent with modelled data from preclinical studies. Here, we show that BWC0977 has the potential to treat a range of critical-care infections including MDR bacterial pneumonias.
Collapse
Affiliation(s)
- Shahul Hameed P
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Harish Kotakonda
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sreevalli Sharma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Radha Nandishaiah
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Nainesh Katagihallimath
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Ranga Rao
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Claire Sadler
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | - Ian Slater
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | - Michael Morton
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | | | - Ed Griffen
- Medchemica Ltd., No. 8162245, Ebenezer House, Newcastle-under-Lyme, Staffordshire, ST5 2BE, England
| | - Dhanashree Pillai
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sambasiva Reddy
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Nagakumar Bharatham
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Suryanarayanan Venkatesan
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Venugopal Jonnalagadda
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Ramesh Jayaraman
- TheraIndx Lifesciences Pvt. Ltd., Sy No. 27, Deganahalli, Bangalore, 562123, India
| | - Mahesh Nanjundappa
- TheraIndx Lifesciences Pvt. Ltd., Sy No. 27, Deganahalli, Bangalore, 562123, India
| | - Maitrayee Sharma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Savitha Raveendran
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sreenath Rajagopal
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Harikrishna Tumma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Amy Watters
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Holly Becker
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Jill Lindley
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Robert Flamm
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Michael Huband
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Dan Sahm
- IHMA USA, 2122 Palmer Drive, Schaumburg, IL, 60173-3817, USA
| | - Meredith Hackel
- IHMA USA, 2122 Palmer Drive, Schaumburg, IL, 60173-3817, USA
| | | | - Ruwanthi Kolamunnage-Dona
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Jennifer Unsworth
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Laura Mcentee
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Nikki Farrington
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Dhanasekaran Manickam
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Narayana Chandrashekara
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Sivakandan Jayachandiran
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Hrushikesava Reddy
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Sathya Shanker
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Vijay Richard
- Narayana Health, Mazumdar Shaw Medical Center, 258/A, Bommasandra Industrial Area, Hosur Road, Bangalore, 560 099, India
| | - Teby Thomas
- Microbiology laboratory, St. John's Hospital, Sarjapur Road, Bangalore, 560 034, India
| | - Savitha Nagaraj
- Microbiology laboratory, St. John's Hospital, Sarjapur Road, Bangalore, 560 034, India
| | - Santanu Datta
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Vasan Sambandamurthy
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Vasanthi Ramachandran
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Robert Clay
- Highbury Regulatory Science Limited, SK10 4TG, Nether Alderley, Cheshire, SK10 4TG, UK
| | - John Tomayko
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Shampa Das
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Balasubramanian V
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India.
| |
Collapse
|
2
|
Peng Y, Zhang AH, Wei L, Welsh WJ. Preclinical Evaluation of Sigma 1 Receptor Antagonists as a Novel Treatment for Painful Diabetic Neuropathy. ACS Pharmacol Transl Sci 2024; 7:2358-2368. [PMID: 39144554 PMCID: PMC11320727 DOI: 10.1021/acsptsci.4c00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024]
Abstract
The global prevalence of diabetes is steadily rising, with an estimated 537 million adults affected by diabetes in 2021, projected to reach 783 million by 2045. A severe consequence of diabetes is the development of painful diabetic neuropathy (PDN), afflicting approximately one in every three diabetic patients and significantly compromising their quality of life. Current pharmacotherapies for PDN provide inadequate pain relief for many patients, underscoring the need for novel treatments that are both safe and effective. The Sigma 1 Receptor (S1R) is a ligand-operated chaperone protein that resides at the mitochondria-associated membrane of the endoplasmic reticulum. The S1R has been shown to play crucial roles in regulating cellular processes implicated in pain modulation. This study explores the potential of PW507, a novel S1R antagonist, as a therapeutic candidate for PDN. PW507 exhibited promising in vitro and in vivo properties in terms of ADME, toxicity, pharmacokinetics, and safety. In preclinical rat models of Streptozotocin-induced diabetic neuropathy, PW507 demonstrated significant efficacy in alleviating mechanical allodynia and thermal hyperalgesia following both acute and chronic (2-week) administration, without inducing tolerance and visual evidence of toxicity. To the best of our knowledge, this is the first report to evaluate an S1R antagonist in STZ-induced diabetic rats following both acute and 2-week chronic administration, offering compelling preclinical evidence for the potential use of PW507 as a promising therapeutic option for PDN.
Collapse
Affiliation(s)
- Youyi Peng
- Biomedical
Informatics Shared Resource, Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903, United States
| | - Allen H. Zhang
- Department
of Biology, Emory College of Arts and Sciences, Atlanta, Georgia 30322, United States
| | - Liping Wei
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - William J. Welsh
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
3
|
Christmann U, Garriga L, Llorente AV, Díaz JL, Pascual R, Bordas M, Dordal A, Porras M, Yeste S, Reinoso RF, Vela JM, Almansa C. WLB-87848, a Selective σ 1 Receptor Agonist, with an Unusually Positioned NH Group as Positive Ionizable Moiety and Showing Neuroprotective Activity. J Med Chem 2024; 67:9150-9164. [PMID: 38753759 DOI: 10.1021/acs.jmedchem.4c00288] [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: 05/18/2024]
Abstract
The synthesis and pharmacological activity of a new series of thieno[2,3-d]pyrimidin-4(3H)-one derivatives as sigma-1 receptor (σ1R) ligands are reported. A hit from a high-throughput screening program was evolved into a highly potent and selective σ1R agonist (14qR) that contains a free NH group as positive ionizable moiety, not fulfilling the usual pharmacophoric features of the σ1R. The compound shows good physicochemical and ADMET characteristics, displays an agonist profile in the binding immunoglobulin protein/σ1R association assay, induces neuron viability in an in vitro model of β-amyloid peptide intoxication, and presents positive results against recognition memory impairment induced by hippocampal injection of Aβ peptide in rats after oral treatment, altogether making 14qR (WLB-87848) an interesting candidate for neuroprotection.
Collapse
Affiliation(s)
- Ute Christmann
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Lourdes Garriga
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Ana Virginia Llorente
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - José Luis Díaz
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Rosalía Pascual
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Magda Bordas
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Albert Dordal
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Mónica Porras
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Sandra Yeste
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Raquel F Reinoso
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - José Miguel Vela
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| | - Carmen Almansa
- Welab Barcelona, Parc Científic Barcelona, C/Baldiri Reixac 4-8, Barcelona 08028, Spain
| |
Collapse
|
4
|
Thoma G, Miltz W, Srinivas H, Penno CA, Kiffe M, Gajewska M, Klein K, Evans A, Beerli C, Röhn TA. Structure-Guided Elaboration of a Fragment-Like Hit into an Orally Efficacious Leukotriene A4 Hydrolase Inhibitor. J Med Chem 2024. [PMID: 38476002 DOI: 10.1021/acs.jmedchem.4c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have provided strong evidence that LTA4H is an attractive drug target for the treatment of chronic inflammatory diseases. Here, we describe the transformation of compound 2, a fragment-like hit, into the potent inhibitor of LTA4H 3. Our strategy involved two key steps. First, we aimed to increase the polarity of fragment 2 to improve its drug-likeness, particularly its solubility, while preserving both its promising potency and low molecular weight. Second, we utilized structural information and incorporated a basic amino function, which allowed for the formation of an essential hydrogen bond with Q136 of LTA4H and consequently enhanced the potency. Compound 3 exhibited exceptional selectivity and showed oral efficacy in a KRN passive serum-induced arthritis model in mice. The anticipated human dose to achieve 90% target engagement at the trough concentration was determined to be 40 mg administered once daily.
Collapse
Affiliation(s)
- Gebhard Thoma
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Wolfgang Miltz
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Honnappa Srinivas
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carlos A Penno
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Kiffe
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Monika Gajewska
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Kai Klein
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Amanda Evans
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Beerli
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Till A Röhn
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| |
Collapse
|
5
|
Yamasaki M, Maki T, Mochida T, Hamada T, Watanabe-Matsumoto S, Konagaya S, Kaneko M, Ito R, Ueno H, Toyoda T. Xenogenic Engraftment of Human-Induced Pluripotent Stem Cell-Derived Pancreatic Islet Cells in an Immunosuppressive Diabetic Göttingen Mini-Pig Model. Cell Transplant 2024; 33:9636897241288932. [PMID: 39401129 PMCID: PMC11489945 DOI: 10.1177/09636897241288932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/21/2024] Open
Abstract
In the development of cell therapy products, immunocompromised animal models closer in size to humans are valuable for enhancing the translatability of in vivo findings to clinical trials. In the present study, we generated immunocompromised type 1 diabetic Göttingen mini-pig models and demonstrated the engraftment of human-induced pluripotent stem cell-derived pancreatic islet cells (iPICs). We induced hyperglycemia with a concomitant reduction in endogenous C-peptide levels in pigs that underwent thymectomy and splenectomy. After estimating the effective in vivo dose of immunosuppressants (ISs) via in vitro testing, we conducted exploratory implantation of iPICs using various implantation methods under IS treatments in one pig. Five weeks after implantation, histological analysis of the implanted iPICs embedded in fibrin gel revealed numerous islet-like structures with insulin-positive cells. Moreover, the area of the insulin-positive cells in the pre-peritoneally implanted grafts was greater than in the subcutaneously implanted grafts. Immunohistochemical analyses further revealed that these iPIC grafts contained cells positive for glucagon, somatostatin, and pancreatic polypeptides, similar to naturally occurring islets. The engraftment of iPICs was successfully reproduced. These data support the observation that the iPICs engrafted well, particularly in the pre-peritoneal space of the newly generated immunocompromised diabetic mini-pigs, forming islet-like endocrine clusters. Future evaluation of human cells in this immunocompromised pig model could accelerate and development of cell therapy products.
Collapse
Affiliation(s)
- Midori Yamasaki
- T-CiRA Discovery and Innovation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
| | | | - Taisuke Mochida
- T-CiRA Discovery and Innovation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
| | - Teruki Hamada
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Japan
| | - Saori Watanabe-Matsumoto
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shuhei Konagaya
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Orizuru Therapeutics, Inc., Fujisawa, Japan
| | - Manami Kaneko
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Japan
| | - Ryo Ito
- T-CiRA Discovery and Innovation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
- Orizuru Therapeutics, Inc., Fujisawa, Japan
| | - Hikaru Ueno
- T-CiRA Discovery and Innovation, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
- Orizuru Therapeutics, Inc., Fujisawa, Japan
| | - Taro Toyoda
- Takeda-CiRA Joint Program for iPS Cell Applications (T-CiRA), Fujisawa, Japan
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Hartl N, Jürgens DC, Carneiro S, König AC, Xiao X, Liu R, Hauck SM, Merkel OM. Protein corona investigations of polyplexes with varying hydrophobicity - From method development to in vitro studies. Int J Pharm 2023; 643:123257. [PMID: 37482228 DOI: 10.1016/j.ijpharm.2023.123257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/11/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
In the field of non-viral drug delivery, polyplexes (PXs) represent an advanced investigated and highly promising tool for the delivery of nucleic acids. Upon encountering physiological fluids, they adsorb biological molecules to form a protein corona (PC), that influence PXs biodistribution, transfection efficiencies and targeting abilities. In an effort to understand protein - PX interactions and the effect of PX material on corona composition, we utilized cationic branched 10 kDa polyethyleneimine (b-PEI) and a hydrophobically modified nylon-3 polymer (NM0.2/CP0.8) within this study to develop appropriate methods for PC investigations. A centrifugation procedure for isolating hard corona - PX complexes (PCPXs) from soft corona proteins after incubating the PXs in fetal bovine serum (FBS) for PC formation was successfully optimized and the identification of proteins by a liquid chromatography-tandem mass spectrometry (LC-MS-MS) method clearly demonstrated that the PC composition is affected by the underlying PXs material. With regard to especially interesting functional proteins, which might be able to induce active targeting effects, several candidates could be detected on b-PEI and NM0.2/CP0.8 PXs. These results are of high interest to better understand how the design of PXs impacts the PC composition and subsequently PCPXs-cell interactions to enable precise adjustment of PXs for targeted drug delivery.
Collapse
Affiliation(s)
- Natascha Hartl
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - David C Jürgens
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Simone Carneiro
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Ann-Christine König
- Metbolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Heidemannsstr. 1, 80939 Munich, Germany
| | - Ximian Xiao
- East China University of Science and Technology, 30 Meilong Rd, Shanghai, China
| | - Runhui Liu
- East China University of Science and Technology, 30 Meilong Rd, Shanghai, China
| | - Stefanie M Hauck
- Metbolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Heidemannsstr. 1, 80939 Munich, Germany
| | - Olivia M Merkel
- Ludwig-Maximilians-University, Pharmaceutical Technology and Biopharmaceutics, Butenandtstr. 5-13, 81377 Munich, Germany.
| |
Collapse
|
7
|
Kang DH, Ahn S, Chae JW, Song JS. Differential effects of two phosphodiesterase 4 inhibitors against lipopolysaccharide-induced neuroinflammation in mice. BMC Neurosci 2023; 24:39. [PMID: 37525115 PMCID: PMC10391911 DOI: 10.1186/s12868-023-00810-7] [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: 03/09/2023] [Accepted: 07/06/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Several phosphodiesterase 4 (PDE4) inhibitors have emerged as potential therapeutics for central nervous system (CNS) diseases. This study investigated the pharmacological effects of two selective PDE4 inhibitors, roflumilast and zatolmilast, against lipopolysaccharide-induced neuroinflammation. RESULTS In BV-2 cells, the PDE4 inhibitor roflumilast reduced the production of nitric oxide and tumor necrosis factor-α (TNF-α) by inhibiting NF-κB phosphorylation. Moreover, mice administered roflumilast had significantly reduced TNF-α, interleukin-1β (IL-1β), and IL-6 levels in plasma and brain tissues. By contrast, zatolmilast, a PDE4D inhibitor, showed no anti-neuroinflammatory effects in vitro or in vivo. Next, in vitro and in vivo pharmacokinetic studies of these compounds in the brain were performed. The apparent permeability coefficients of 3 µM roflumilast and zatolmilast were high (> 23 × 10-6 cm/s) and moderate (3.72-7.18 × 10-6 cm/s), respectively, and increased in a concentration-dependent manner in the MDR1-MDCK monolayer. The efflux ratios were < 1.92, suggesting that these compounds are not P-glycoprotein substrates. Following oral administration, both roflumilast and zatolmilast were slowly absorbed and eliminated, with time-to-peak drug concentrations of 2-2.3 h and terminal half-lives of 7-20 h. Assessment of their brain dispositions revealed the unbound brain-to-plasma partition coefficients of roflumilast and zatolmilast to be 0.17 and 0.18, respectively. CONCLUSIONS These findings suggest that roflumilast, but not zatolmilast, has the potential for use as a therapeutic agent against neuroinflammatory diseases.
Collapse
Affiliation(s)
- Dong Ho Kang
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Sunjoo Ahn
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea
| | - Jung Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Jin Sook Song
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea.
| |
Collapse
|
8
|
Tavakoli Hafshajani K, Sohrabi N, Eslami Moghadam M, Oftadeh M. Spectroscopy and molecular dynamic study of the interaction of calf thymus DNA by anticancer Pt complex with butyl glycine ligand. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 299:122826. [PMID: 37216815 DOI: 10.1016/j.saa.2023.122826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Despite the past few decades since the discovery of anticancer drugs, there is still no definitive treatment for its treatment. Cisplatin is a chemotherapy medication used to treat some cancers. In this research, the DNA binding affinity of Pt complex with butyl glycine ligand was studied by various spectroscopy methods and simulation studies. Fluorescence and UV-Vis spectroscopic data showed groove binding in ct-DNA-[Pt(NH3)2(butylgly)]NO3 complex formation by the spontaneous process. The results were also confirmed by small changes in CD spectra and thermal study (Tm), as well as the quenching emission of [Pt(NH3)2(butylgly)]NO3 complex on DNA. Finally, thermodynamic and binding parameters displayed that hydrophobic forces are the main forces. Based on docking simulation, [Pt(NH3)2(butylgly)]NO3 could bind to DNA and via minor groove binding on C-G center on DNA, formed a stable DNA complex.
Collapse
Affiliation(s)
| | - Nasrin Sohrabi
- Department of Chemistry, Payame Noor University (PNU), P.O.Box 19395-4697, Tehran, Iran.
| | | | - Mohsen Oftadeh
- Department of Chemistry, Payame Noor University (PNU), P.O.Box 19395-4697, Tehran, Iran
| |
Collapse
|
9
|
Dilshad M, Shah A, Munir S. Electroanalysis of Ibuprofen and Its Interaction with Bovine Serum Albumin. Molecules 2022; 28:49. [PMID: 36615246 PMCID: PMC9821973 DOI: 10.3390/molecules28010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
The current work presents a sensitive, selective, cost-effective, and environmentally benign protocol for the detection of ibuprofen (IBP) by an electrochemical probe made of a glassy carbon electrode modified with Ag-ZnO and MWCNTs. Under optimized conditions, the designed sensing platform was found to sense IBP up to a 28 nM limit of detection. The interaction of IBP with bovine serum albumin (BSA) was investigated by differential pulse voltammetry. IBP-BSA binding parameters such as the binding constant and the stoichiometry of complexation were calculated. The results revealed that IBP and BSA form a single strong complex with a binding constant value of 8.7 × 1013. To the best of our knowledge, this is the first example that reports not only IBP detection but also its BSA complexation.
Collapse
Affiliation(s)
- Muhammad Dilshad
- Department of Chemistry, Quaid-Azam University, Islamabad 45320, Pakistan
| | - Afzal Shah
- Department of Chemistry, Quaid-Azam University, Islamabad 45320, Pakistan
| | - Shamsa Munir
- School of Applied Sciences and Humanities, National University of Technology (NUTECH), Islamabad 44000, Pakistan
| |
Collapse
|
10
|
Tavakoli Hafshejani K, Sohrabi N, Eslami Moghadam M, Oftadeh M. Investigation of the physico-chemical interaction of ct-DNA with Anticancer Glycine Derivative of Pt-complex by applying docking and MD simulation methods and multi-spectroscopic techniques. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
11
|
Loryan I, Reichel A, Feng B, Bundgaard C, Shaffer C, Kalvass C, Bednarczyk D, Morrison D, Lesuisse D, Hoppe E, Terstappen GC, Fischer H, Di L, Colclough N, Summerfield S, Buckley ST, Maurer TS, Fridén M. Unbound Brain-to-Plasma Partition Coefficient, K p,uu,brain-a Game Changing Parameter for CNS Drug Discovery and Development. Pharm Res 2022; 39:1321-1341. [PMID: 35411506 PMCID: PMC9246790 DOI: 10.1007/s11095-022-03246-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/22/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE More than 15 years have passed since the first description of the unbound brain-to-plasma partition coefficient (Kp,uu,brain) by Prof. Margareta Hammarlund-Udenaes, which was enabled by advancements in experimental methodologies including cerebral microdialysis. Since then, growing knowledge and data continue to support the notion that the unbound (free) concentration of a drug at the site of action, such as the brain, is the driving force for pharmacological responses. Towards this end, Kp,uu,brain is the key parameter to obtain unbound brain concentrations from unbound plasma concentrations. METHODS To understand the importance and impact of the Kp,uu,brain concept in contemporary drug discovery and development, a survey has been conducted amongst major pharmaceutical companies based in Europe and the USA. Here, we present the results from this survey which consisted of 47 questions addressing: 1) Background information of the companies, 2) Implementation, 3) Application areas, 4) Methodology, 5) Impact and 6) Future perspectives. RESULTS AND CONCLUSIONS From the responses, it is clear that the majority of the companies (93%) has established a common understanding across disciplines of the concept and utility of Kp,uu,brain as compared to other parameters related to brain exposure. Adoption of the Kp,uu,brain concept has been mainly driven by individual scientists advocating its application in the various companies rather than by a top-down approach. Remarkably, 79% of all responders describe the portfolio impact of Kp,uu,brain implementation in their companies as 'game-changing'. Although most companies (74%) consider the current toolbox for Kp,uu,brain assessment and its validation satisfactory for drug discovery and early development, areas of improvement and future research to better understand human brain pharmacokinetics/pharmacodynamics translation have been identified.
Collapse
Affiliation(s)
- Irena Loryan
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, Sweden.
| | | | - Bo Feng
- DMPK, Vertex Pharmaceuticals, Boston, Massachusetts, 02210, USA
| | | | - Christopher Shaffer
- External Innovation, Research & Development, Biogen Inc., Cambridge, Massachusetts, USA
| | - Cory Kalvass
- DMPK-BA, AbbVie, Inc., North Chicago, Illinois, USA
| | - Dallas Bednarczyk
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | | | | | - Edmund Hoppe
- DMPK, Boehringer-Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Holger Fischer
- Translational PK/PD and Clinical Pharmacology, Pharmaceutical Sciences, Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut, USA
| | | | - Scott Summerfield
- Bioanalysis Immunogenicity and Biomarkers, GSK, Gunnels Wood Road, Stevenage, SG1 2NY, Hertfordshire, UK
| | | | - Tristan S Maurer
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, USA
| | - Markus Fridén
- Department of Pharmacy, Uppsala University, Box 580, Uppsala, Sweden
- Inhalation Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
12
|
Namazi N. A Modified Polymeric Nano-formulation to Control Binding and Release of Insulin. J Pharm Sci 2022; 111:2481-2489. [DOI: 10.1016/j.xphs.2022.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
13
|
Fagerholm U, Spjuth O, Hellberg S. Comparison between lab variability and in silico prediction errors for the unbound fraction of drugs in human plasma. Xenobiotica 2021; 51:1095-1100. [PMID: 34346291 DOI: 10.1080/00498254.2021.1964044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Variability of the unbound fraction in plasma (fu) between labs, methods and conditions is known to exist. Variability and uncertainty of this parameter influence predictions of the overall pharmacokinetics of drug candidates and might jeopardise safety in early clinical trials. Objectives of this study were to evaluate the variability of human in vitro fu-estimates between labs for a range of different drugs, and to develop and validate an in silico fu-prediction method and compare the results to the lab variability.A new in silico method with prediction accuracy (Q2) of 0.69 for log fu was developed. The median and maximum prediction errors were 1.9- and 92-fold, respectively. Corresponding estimates for lab variability (ratio between max and min fu for each compound) were 2.0- and 185-fold, respectively. Greater than 10-fold lab variability was found for 14 of 117 selected compounds.Comparisons demonstrate that in silico predictions were about as reliable as lab estimates when these have been generated during different conditions. Results propose that the new validated in silico prediction method is valuable not only for predictions at the drug design stage, but also for reducing uncertainties of fu-estimations and improving safety of drug candidates entering the clinical phase.
Collapse
Affiliation(s)
| | - Ola Spjuth
- Prosilico AB, Huddinge, Sweden.,Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
14
|
A novel insight into the cytotoxic effects of Tephrosin with calf thymus DNA: Experimental and in silico approaches. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2020.114728] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
15
|
Seyfinejad B, Khoubnasabjafari M, Ziaei SE, Ozkan SA, Jouyban A. Electromembrane extraction as a new approach for determination of free concentration of phenytoin in plasma using capillary electrophoresis. ACTA ACUST UNITED AC 2020; 28:615-624. [PMID: 32803689 DOI: 10.1007/s40199-020-00366-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/05/2020] [Indexed: 01/26/2023]
Abstract
PURPOSE Electromembrane extraction is a new membrane-based extraction method in which charged compounds are extracted by an electric field. So far, this method has been used to extract and isolate a variety of acidic and basic drugs from various samples, including blood and plasma. However, in this procedure, it is not yet clear whether only unbound fraction of a drug is extracted or the total drug. The aim of this study is to reveal the nature of drug extraction in the presence of plasma proteins. METHODS To determine the nature of the extraction, the electromembrane extraction was performed from plasma solutions of phenytoin with concentrations 0.03 and 1.0 μg/mL, then the result was compared with the values obtained from the electromembrane extraction of ultrafiltrate of the same solutions (free concentration) and protein-free ultrafiltrate of plasma with final concentration of 0.03 and 1.0 μg/mL (total concentration). For this purpose, EME followed by capillary electrophoresis coupled with diode array detection was optimized and validated. RESULTS The results showed that the electromembrane extraction method was only able to extract the unbound fraction of phenytoin from plasma samples. The method was validated over a concentration range of 0.03-4 μg/mL. The inter and intra-assay precisions were less than 6.7%. The phenytoin protein binding was also determined to be in agreement with the literature data and confirms the validity of this method. CONCLUSION This sensitive and quick EME approach for determining the free concentration of a phenytoin, can be a good alternative to classic methods for therapeutic drug monitoring and pharmacokinetic studies.
Collapse
Affiliation(s)
- Behrouz Seyfinejad
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Khoubnasabjafari
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Eivaz Ziaei
- Neurosciences Research Center, Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sibel A Ozkan
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, 06100, Ankara, Turkey
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. .,Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Quantitative Microdialysis: Experimental Protocol and Software for Small Molecule Protein Affinity Determination and for Exclusion of Compounds with Poor Physicochemical Properties. Methods Protoc 2020; 3:mps3030055. [PMID: 32751503 PMCID: PMC7563421 DOI: 10.3390/mps3030055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/03/2022] Open
Abstract
Quantitative microdialysis is a traditional biophysical affinity determination technique. In the development of the detailed experimental protocol presented, we used commercially available equipment, rapid equilibrium dialysis (RED) devices (ThermoFisher Scientific), which means that it is open to most laboratories. The target protein and test compound are incubated in a chamber partitioned to allow only small molecules to transition to a larger reservoir chamber, then reversed-phase high performance liquid chromatography (RP-HPLC) or liquid chromatography–mass spectrometry (LC–MS) is used to determine the abundance of compound in each chamber. A higher compound concentration measured in the chamber that contains the target protein indicates binding. As a novel, and differentiating contribution, we present a protocol for mathematical analysis of experimental data. We provide the equations and the software to yield dissociation constants for the test compound-target protein complex up to 0.5 mM KD, and we quantitatively discuss the limitations of affinities in relation to measured compound concentrations.
Collapse
|
17
|
Moein MM, Halldin C. Sample preparation techniques for protein binding measurement in radiopharmaceutical approaches: A short review. Talanta 2020; 219:121220. [PMID: 32887121 DOI: 10.1016/j.talanta.2020.121220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 10/24/2022]
Abstract
Plasma protein binding (PPB) measurement is a key step in radiopharmaceutical studies for the development of positron emission tomography (PET) radioligands. PPB refers to the binding degree of a radioligand, radiotracer, or drug to blood plasma proteins or tissues after administration into the body. Several techniques have been successfully developed and applied for PPB measurement of PET radioligands. However, there is room for progress among these techniques in relation to duration time, adaptability with nonpolar radioligands, in vivo measurement, specificity, and selectivity. This mini review gives a brief overview of advances, limitations, and prospective applications of commercially-available PPB methods.
Collapse
Affiliation(s)
- Mohammad Mahdi Moein
- Karolinska Radiopharmacy, Karolinska University Hospital, S-171 64 Stockholm, Sweden; Karolinska Institutet, Department of Oncology-Pathology, J5:20, S-171 77 Stockholm, Sweden.
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| |
Collapse
|
18
|
Wernevik J, Bergström F, Novén A, Hulthe J, Fredlund L, Addison D, Holmgren J, Strömstedt PE, Rehnström E, Lundbäck T. A Fully Integrated Assay Panel for Early Drug Metabolism and Pharmacokinetics Profiling. Assay Drug Dev Technol 2020; 18:157-179. [PMID: 32407132 PMCID: PMC7567642 DOI: 10.1089/adt.2020.970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Evaluation and optimization of physicochemical and metabolic properties of compounds are a crucial component of the drug development process. Continuous access to this information during the design-make-test-analysis cycle enables identification of chemical entities with suitable properties for efficient project progression. In this study, we describe an integrated and automated assay panel (DMPK Wave 1) that informs weekly on lipophilicity, solubility, human plasma protein binding, and metabolic stability in rat hepatocytes and human liver microsomes. All assays are running in 96-well format with ultraperformance liquid chromatography-mass spectrometry (MS)/MS as read-out. A streamlined overall workflow has been developed by optimizing all parts of the process, including shipping of compounds between sites, use of fit-for-purpose equipment and information systems, and technology for compound requesting, data analysis, and reporting. As a result, lead times can be achieved that well match project demands across sites independently of where compounds are synthesized. This robust screening strategy is run on a weekly basis and enables optimization of structure-activity relationships in parallel with DMPK properties to allow efficient and informed decision making.
Collapse
Affiliation(s)
- Johan Wernevik
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Bergström
- DMPK, Early CVRM, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Novén
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Johan Hulthe
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linda Fredlund
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Dan Addison
- Sample Management, Discovery Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | - Jan Holmgren
- Sample Management, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Per-Erik Strömstedt
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Erika Rehnström
- Clinical Sampling & Alliances, Precision Medicine, AstraZeneca, Gothenburg, Sweden
| | - Thomas Lundbäck
- Mechanistic Biology & Profiling, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
19
|
Ravelli D, Isernia P, Acquarulo A, Profumo A, Merli D. Voltammetric Determination of Binding Constant and Stoichiometry of Albumin (Human, Bovine, Ovine)–Drug Complexes. Anal Chem 2019; 91:10110-10115. [DOI: 10.1021/acs.analchem.9b02088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Davide Ravelli
- Università degli Studi di Pavia, Dipartimento di Chimica, Viale Taramelli 12, 27100 Pavia, Italia
| | - Paola Isernia
- Fondazione IRCCS Policlinico San Matteo, Servizio di Immunoematologia e Medicina Trasfusionale, Centro Lavorazione e Validazione Emocomponenti (CLV), Viale Camillo Golgi 19, 27100 Pavia, Italia
| | - Andrea Acquarulo
- Università degli Studi di Pavia, Dipartimento di Chimica, Viale Taramelli 12, 27100 Pavia, Italia
| | - Antonella Profumo
- Università degli Studi di Pavia, Dipartimento di Chimica, Viale Taramelli 12, 27100 Pavia, Italia
| | - Daniele Merli
- Università degli Studi di Pavia, Dipartimento di Chimica, Viale Taramelli 12, 27100 Pavia, Italia
| |
Collapse
|
20
|
Ferguson KC, Luo YS, Rusyn I, Chiu WA. Comparative analysis of Rapid Equilibrium Dialysis (RED) and solid phase micro-extraction (SPME) methods for In Vitro-In Vivo extrapolation of environmental chemicals. Toxicol In Vitro 2019; 60:245-251. [PMID: 31195086 DOI: 10.1016/j.tiv.2019.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/15/2019] [Accepted: 06/08/2019] [Indexed: 01/17/2023]
Abstract
In vitro to in vivo extrapolation (IVIVE) is a critical component of the efforts to prioritize and assess environmental chemicals using high-throughput in vitro assays. The plasma unbound fraction (Fub) is a key toxicokinetic parameter in IVIVE, and is usually measured via the Rapid Equilibrium Dialysis (RED) assay widely used for pharmaceuticals. However, pharmaceuticals have a narrower range of physicochemical properties than environmental chemicals. Motivated by the observation that high LogKOW compounds appeared to have disproportionately low Fub measurements using RED, we added a protein-free control in order to verify equilibration to 100% unbound in the absence of proteins. We found that many high LogKOW non-pharmaceuticals fail to equilibrate in RED in protein-free controls, and thus had apparent values of Fub = 0 in plasma. In these cases, Solid Phase Microextraction (SPME) as an alternative method provided an accurate, though more time-consuming, alternative to accurately determine Fub. We propose an updated IVIVE workflow that adds a protein-free control to the RED protocol, with the use of alternative approaches, such as SPME, in cases where compounds fail to adequately equilibrate. These refinements will provide additional confidence in the use of IVIVE as part of high-throughput screening programs of chemicals.
Collapse
Affiliation(s)
- Kyle C Ferguson
- Department of Veterinary Integrative Biosciences, College of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Yu-Syuan Luo
- Department of Veterinary Integrative Biosciences, College of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, College of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
21
|
Sasikumar PG, Ramachandra RK, Adurthi S, Dhudashiya AA, Vadlamani S, Vemula K, Vunnum S, Satyam LK, Samiulla DS, Subbarao K, Nair R, Shrimali R, Gowda N, Ramachandra M. A Rationally Designed Peptide Antagonist of the PD-1 Signaling Pathway as an Immunomodulatory Agent for Cancer Therapy. Mol Cancer Ther 2019; 18:1081-1091. [PMID: 31015307 DOI: 10.1158/1535-7163.mct-18-0737] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/26/2018] [Accepted: 04/15/2019] [Indexed: 11/16/2022]
Abstract
Pioneering success of antibodies targeting immune checkpoints such as PD-1 and CTLA4 has opened novel avenues for cancer immunotherapy. Along with impressive clinical activity, severe immune-related adverse events (irAE) due to the breaking of immune self-tolerance are becoming increasingly evident in antibody-based approaches. As a strategy to better manage severe adverse effects, we set out to discover an antagonist targeting PD-1 signaling pathway with a shorter pharmacokinetic profile. Herein, we describe a peptide antagonist NP-12 that displays equipotent antagonism toward PD-L1 and PD-L2 in rescue of lymphocyte proliferation and effector functions. In preclinical models of melanoma, colon cancer, and kidney cancers, NP-12 showed significant efficacy comparable with commercially available PD-1-targeting antibodies in inhibiting primary tumor growth and metastasis. Interestingly, antitumor activity of NP-12 in a preestablished CT26 model correlated well with pharmacodynamic effects as indicated by intratumoral recruitment of CD4 and CD8 T cells, and a reduction in PD-1+ T cells (both CD4 and CD8) in tumor and blood. In addition, NP-12 also showed additive antitumor activity in preestablished tumor models when combined with tumor vaccination or a chemotherapeutic agent such as cyclophosphamide known to induce "immunologic cell death." In summary, NP-12 is the first rationally designed peptide therapeutic targeting PD-1 signaling pathways exhibiting immune activation, excellent antitumor activity, and potential for better management of irAEs.
Collapse
Affiliation(s)
| | | | - Srinivas Adurthi
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | - Amit A Dhudashiya
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | | | | | | | - Leena K Satyam
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | | | | | - Rashmi Nair
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | - Rajeev Shrimali
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | - Nagaraj Gowda
- Aurigene Discovery Technologies Limited, Bangalore, Karnataka, India
| | | |
Collapse
|
22
|
Smith SA, Waters NJ. Pharmacokinetic and Pharmacodynamic Considerations for Drugs Binding to Alpha-1-Acid Glycoprotein. Pharm Res 2018; 36:30. [PMID: 30593605 PMCID: PMC7089466 DOI: 10.1007/s11095-018-2551-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022]
Abstract
According to the free drug hypothesis only the unbound drug is available to act at physiological sites of action, and as such the importance of plasma protein binding primarily resides in its impact on pharmacokinetics and pharmacodynamics. Of the major plasma proteins, alpha-1-acid glycoprotein (AAG) represents an intriguing one primarily due to the high affinity, low capacity properties of this protein. In addition, there are marked species and age differences in protein expression, homology and drug binding affinity. As such, a thorough understanding of drug binding to AAG can help aid and improve the translation of pharmacokinetic/pharmacodynamic (PK/PD) relationships from preclinical species to human as well as adults to neonates. This review provides a comprehensive overview of our current understanding of the biochemistry of AAG; endogenous function, impact of disease, utility as a biomarker, and impact on PK/PD. Experimental considerations are discussed as well as recommendations for understanding the potential impact of AAG on PK through drug discovery and early development.
Collapse
Affiliation(s)
- Sherri A Smith
- Drug Metabolism, Pharmacokinetics and Bioanalytical, H3 Biomedicine, 300 Technology Square, Cambridge, Massachusetts, 02139, USA.
| | - Nigel J Waters
- Nonclinical Development, Relay Therapeutics, 215 First Street, Cambridge, Massachusetts, USA
| |
Collapse
|
23
|
Badée J, Qiu N, Parrott N, Collier AC, Schmidt S, Fowler S. Optimization of Experimental Conditions of Automated Glucuronidation Assays in Human Liver Microsomes Using a Cocktail Approach and Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry. Drug Metab Dispos 2018; 47:124-134. [PMID: 30478159 DOI: 10.1124/dmd.118.084301] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/20/2018] [Indexed: 11/22/2022] Open
Abstract
UDP-glucuronosyltransferase (UGT)-mediated metabolism is possibly the most important conjugation reaction for marketed drugs. However, there are currently no generally accepted standard incubation conditions for UGT microsomal assays, and substantial differences in experimental design and methodology between laboratories hinder cross-study comparison of in vitro activities. This study aimed to define optimal experimental conditions to determine glucuronidation activity of multiple UGT isoforms simultaneously using human liver microsomes. Hepatic glucuronidation activities of UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, UGT2B4, UGT2B7, UGT2B10, UGT2B15, and UGT2B17 were determined using cocktail incubations of 10 UGT probe substrates. Buffer components and cosubstrates were assessed over a range of concentrations including magnesium chloride (MgCl2; 0-10 mM) and uridine 5'-diphosphoglucuronic acid (UDPGA; 1-25 mM) with either Tris-HCl or potassium phosphate buffer (100 mM, pH 7.4). Greater microsomal glucuronidation activity by different hepatic UGT isoforms was obtained using 10 mM MgCl2 and 5 mM UDPGA with 100 mM Tris-HCl buffer. The influence of bovine serum albumin (BSA; 0.1%-2% w/v) on glucuronidation activity was also assessed. Enzyme- and substrate-dependent effects of BSA were observed, resulting in decreased total activity of UGT1A1, UGT1A3, and UGT2B17 and increased total UGT1A9 and UGT2B7 activity. The inclusion of BSA did not significantly reduce the between-subject variability of UGT activity. Future in vitro UGT profiling studies under the proposed optimized experimental conditions would allow high-quality positive control data to be generated across laboratories, with effective control of a high degree of between-donor variability for UGT activity and for chemical optimization toward lower-clearance drug molecules in a pharmaceutical drug discovery setting.
Collapse
Affiliation(s)
- Justine Badée
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Nahong Qiu
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Neil Parrott
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Abby C Collier
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| | - Stephen Fowler
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, University of Florida at Lake Nona, Orlando, Florida (J.B., S.S.); Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland (N.Q., N.P., S.F.); and Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (A.C.C.)
| |
Collapse
|
24
|
Differences in peritoneal solute transport rates in peritoneal dialysis. Clin Exp Nephrol 2018; 23:122-134. [DOI: 10.1007/s10157-018-1611-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
|
25
|
Frontal analysis capillary electrophoresis: recent advances and future perspectives. Bioanalysis 2018; 10:1143-1159. [DOI: 10.4155/bio-2018-0051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The developments on frontal analysis capillary electrophoresis (FACE) from 2012 to 2017 to study interactions of simple and complex systems are reviewed. Most research papers focused on therapeutic drug-related studies; however, other studies include chemical sensing, drug delivery, inhibitor screening and capillary coating. New ligand–substrate systems such as template-molecularly imprinted polymer systems were reported. Comparison of FACE with other analytical techniques used to investigate binding interaction, and the determination of binding parameters using different isotherm models are also covered. In 2017, eight research papers were reported including new detection by ESI–MS. Future research direction of FACE may include high sensitivity detection and throughput screening of drugs, natural products and biomarkers for clinical diagnosis.
Collapse
|
26
|
Herbst ZM, Shibata S, Fan E, Gelb MH. An Inexpensive, In-House-Made, Microdialysis Device for Measuring Drug-Protein Binding. ACS Med Chem Lett 2018. [PMID: 29541374 DOI: 10.1021/acsmedchemlett.7b00316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An inexpensive, in-house made microdialysis device is described that is suitable for measuring the binding of small molecules including drug candidates to serum proteins or other macromolecules. The device is based on the standard equilibrium dialysis method to measure the fraction of low molecular weight compound bound to proteins. It is constructed from a standard polypropylene 96-well plate, dialysis tubing, and low viscosity epoxy resin. The device can be readily prepared for a small fraction of the cost of a commercial, multichamber microdialysis device. Drug-protein binding results are provided, which validates the device.
Collapse
Affiliation(s)
- Zackary M. Herbst
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Sayaka Shibata
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Erkang Fan
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Michael H. Gelb
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
27
|
Industry Perspective on Contemporary Protein-Binding Methodologies: Considerations for Regulatory Drug-Drug Interaction and Related Guidelines on Highly Bound Drugs. J Pharm Sci 2017; 106:3442-3452. [DOI: 10.1016/j.xphs.2017.09.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/07/2017] [Indexed: 11/21/2022]
|
28
|
Sun L, Yang H, Li J, Wang T, Li W, Liu G, Tang Y. In Silico Prediction of Compounds Binding to Human Plasma Proteins by QSAR Models. ChemMedChem 2017; 13:572-581. [DOI: 10.1002/cmdc.201700582] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/18/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Lixia Sun
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Jie Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Tianduanyi Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy; East China University of Science and Technology; Shanghai 200237 China
| |
Collapse
|
29
|
Abstract
Perhaps because they are such commonly used tools, many researchers view antibodies one-dimensionally: Antibody Y binds antigen X. Although few techniques require a comprehensive understanding of any particular antibody's characteristics, well-executed experiments do require a basic appreciation of what is known and, equally as important, what is not known about the antibody being used. Ignorance of the relevant antibody characteristics critical for a particular assay can easily lead to loss of precious resources (time, money, and limiting amounts of sample) and, in worst-case scenarios, erroneous conclusions. Here, we describe various antibody characteristics to provide a more well-rounded perspective of these critical reagents. With this information, it will be easier to make informed decisions on how best to choose and use the available antibodies, as well as knowing when it is essential and how to determine a particular as yet-undefined characteristic.
Collapse
|
30
|
Monitoring drug–serum protein interactions for early ADME prediction through Surface Plasmon Resonance technology. J Pharm Biomed Anal 2017; 144:188-194. [DOI: 10.1016/j.jpba.2017.03.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/03/2017] [Accepted: 03/26/2017] [Indexed: 12/16/2022]
|
31
|
Jafari F, Samadi S, Nowroozi A, Sadrjavadi K, Moradi S, Ashrafi-Kooshk MR, Shahlaei M. Experimental and computational studies on the binding of diazinon to human serum albumin. J Biomol Struct Dyn 2017; 36:1490-1510. [DOI: 10.1080/07391102.2017.1329096] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Fataneh Jafari
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Setareh Samadi
- Department of Toxicology, Shahreza Branch, Islamic Azad University, Shaahreza, Iran
| | - Amin Nowroozi
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Komail Sadrjavadi
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mohsen Shahlaei
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
32
|
Jafari F, Moradi S, Nowroozi A, Sadrjavadi K, Hosseinzadeh L, Shahlaei M. Exploring the binding mechanism of paraquat to DNA by a combination of spectroscopic, cellular uptake, molecular docking and molecular dynamics simulation methods. NEW J CHEM 2017. [DOI: 10.1039/c7nj01645j] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The possibility that paraquat may exert its toxicity also by interaction with DNA is studied using a combination of different computational and experimental techniques.
Collapse
Affiliation(s)
- Fataneh Jafari
- Pharmaceuticas Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, School of Pharmacy, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| | - Amin Nowroozi
- Pharmaceuticas Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| | - Komail Sadrjavadi
- Pharmaceuticas Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| | - Leila Hosseinzadeh
- Pharmaceuticas Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| | - Mohsen Shahlaei
- Medical Biology Research Center, Kermanshah University of Medical Sciences
- Kermanshah
- Iran
| |
Collapse
|
33
|
Molecular insight into the Grandivitin- matrix metalloproteinase 9 interactions. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 162:493-499. [PMID: 27454459 DOI: 10.1016/j.jphotobiol.2016.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 07/16/2016] [Indexed: 11/23/2022]
Abstract
Grandivitin (GRA), a natural coumarin, can inhibit Matrix metalloproteinase 9 (MMP9). Binding characteristics are therefore of interest for pharmacodynamics of GRA and coumarin derivatives. A combination of spectroscopic methods and molecular modeling techniques was used to characterize interaction of GRA with MMP9. Fluorescence spectroscopy showed that GRA could quench the MMP9 fluorescence spectra. Changes in the UV-Vis and FT-IR spectra were observed upon ligand binding along with a significant degree of tryptophan fluorescence quenching on complex formation. Fluorescence studies showed that GRA has an ability to quench the intrinsic fluorescence of MMP9. Molecular modeling analysis showed that GRA to be bound in the large hydrophobic cavity of MMP9. Further investigation of the binding site of GRA within the MMP9 molecule suggested that hydrophobic contacts, hydrogen bond formation and electrostatic interactions account for the binding of GRA. According molecular dynamics (MD) simulation results the ligand can interact with the protein, with affecting the secondary structure of MMP9 and with a modification of its tertiary structure. The biological significance of this work is evident because MMP9 serves as a potential target protein for anticancer agents. The binding study of GRA with MMP9 is of great importance in pharmacy, pharmacology and biochemistry. This work can provide some key data to clinical research and supply the theoretical basis for the new drug candidate designing.
Collapse
|
34
|
Holm KMD, Linnet K. Determination of the unbound fraction of R- and S-methadone in human brain. Int J Legal Med 2016; 130:1519-1526. [DOI: 10.1007/s00414-016-1365-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/29/2016] [Indexed: 10/22/2022]
|
35
|
Mu Y, Liu Y, Xiang J, Zhang Q, Zhai S, Russo DP, Zhu H, Bai X, Yan B. From fighting depression to conquering tumors: a novel tricyclic thiazepine compound as a tubulin polymerization inhibitor. Cell Death Dis 2016; 7:e2143. [PMID: 26986511 PMCID: PMC4823954 DOI: 10.1038/cddis.2016.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/17/2016] [Accepted: 02/17/2016] [Indexed: 12/17/2022]
Abstract
A novel tricyclic thiazepine derivative, 6-(p-tolyl)benzo[f] pyrido[2,3-b][1,4] thiazepine 11,11-dioxide (TBPT), exhibits potent inhibitory effects in two non-small-cell lung cancer cell lines, H460 and its drug-resistant variant, H460TaxR, while exhibiting much less toxic effects on normal human fibroblasts. After five injections of TBPT at a dose of 60 mg/kg, it inhibits H460TaxR tumor growth in xenografted mouse models by 66.7% without causing observable toxicity to normal tissues. Based on gene perturbation data and a series of investigations, we reveal that TBPT is not a P-glycoprotein substrate and it inhibits microtubule formation by targeting tubulin, thereby causing cell cycle arrest at the G2/M stage and eventually inducing apoptosis. This redeployment of anti-depressant compound scaffold for anticancer applications provides a promising future for conquering drug-resistant tumors with fewer side effects.
Collapse
Affiliation(s)
- Y Mu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - Y Liu
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - J Xiang
- The Center for Combinatorial Chemistry and Drug Discovery of Jilin University, The School of Pharmaceutical Sciences and The College of Chemistry, Jilin University, Changchun, Jilin, China
| | - Q Zhang
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - S Zhai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| | - D P Russo
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA
| | - H Zhu
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ, USA.,Department of Chemistry, Rutgers University, Camden, NJ, USA
| | - X Bai
- The Center for Combinatorial Chemistry and Drug Discovery of Jilin University, The School of Pharmaceutical Sciences and The College of Chemistry, Jilin University, Changchun, Jilin, China
| | - B Yan
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, China
| |
Collapse
|
36
|
Exploring binding properties of sertraline with human serum albumin: Combination of spectroscopic and molecular modeling studies. Chem Biol Interact 2015; 242:235-46. [DOI: 10.1016/j.cbi.2015.10.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Revised: 09/25/2015] [Accepted: 10/07/2015] [Indexed: 12/20/2022]
|
37
|
Shokoohinia Y, Gheibi S, Kiani A, Sadrjavadi K, Nowroozi A, Shahlaei M. Multi-spectroscopic and molecular modeling investigation of the interactions between prantschimgin and matrix metalloproteinase 9 (MMP9). LUMINESCENCE 2015; 31:587-593. [PMID: 26311532 DOI: 10.1002/bio.2999] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/09/2015] [Accepted: 07/11/2015] [Indexed: 12/22/2022]
Abstract
The binding of prantschimgin (PRAN) to matrix metalloproteinase 9 (MMP9) was investigated using multiple techniques. Fluorescence spectroscopy showed that PRAN could quench the MMP9 fluorescence spectra. Changes in the UV/vis and Fourier transform infrared (FTIR) spectra were observed upon ligand binding, along with a significant degree of tryptophan fluorescence quenching on complex formation. The interaction of PRAN with MMP9 has also been studied using molecular docking and molecular dynamics (MD) simulation. The binding models demonstrated aspects of PRAN's conformation, active site interaction, important amino acids and hydrogen bonding. Computational mapping of the possible binding site of PRAN revealed that the ligand is bound in a large hydrophobic cavity of MMP9. The MD simulation results suggested that this ligand can interact with the protein, with little affecting the secondary structure. The results not only lead to a better understanding of interactions between PRAN and MMP9, but also provide useful data about the influence of PRAN on the structural conformation. The data provided in this study will be useful for designing a new agonist of MMP9 with the desired activity.
Collapse
Affiliation(s)
- Yalda Shokoohinia
- Novel Drug Delivery Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shayesteh Gheibi
- Student Research Committee School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Kiani
- Pharmacology and Toxicology Department, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Komail Sadrjavadi
- Novel Drug Delivery Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Nowroozi
- Nano Drug Delivery Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
38
|
Kiani A, Almasi K, Shokoohinia Y, Sadrjavadi K, Nowroozi A, Shahlaei M. Combined spectroscopy and molecular modeling studies on the binding of galbanic acid and MMP9. Int J Biol Macromol 2015; 81:308-15. [PMID: 26253511 DOI: 10.1016/j.ijbiomac.2015.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/30/2015] [Accepted: 08/03/2015] [Indexed: 11/16/2022]
Abstract
The molecular mechanism of galbanic acid (GBA) binding to matrix metalloproteinase 9 (MMP9) was investigated by fluorescence quenching, absorption spectroscopy, FT-IR, molecular docking and molecular dynamics (MD) simulation procedures. The fluorescence emission of MMP9 was quenched by GBA. The titration of MMP9 by various amount of GBA was also followed by UV-Vis absorption spectroscopy. The results revealed that GBA, as a biologically active sesquiterpene coumarin derivative, has an ability to bind strongly to MMP9. Molecular docking results indicated that the main active binding site for GBA has been located in a hydrophobic cavity in the vicinity of Zn atom. Moreover, MD simulation results suggested that GBA as a coumarin derivative can interact with MMP9, without affecting the secondary structure of MMP9. MD simulations, molecular docking as computational methods from one hand and experimental data from other hand reciprocally supported each other.
Collapse
Affiliation(s)
- Amir Kiani
- Department of Toxicology and pharmacology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khadijeh Almasi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yalda Shokoohinia
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Komail Sadrjavadi
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Nowroozi
- Nano Drug Delivery Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
39
|
Lambrinidis G, Vallianatou T, Tsantili-Kakoulidou A. In vitro, in silico and integrated strategies for the estimation of plasma protein binding. A review. Adv Drug Deliv Rev 2015; 86:27-45. [PMID: 25819487 DOI: 10.1016/j.addr.2015.03.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/11/2015] [Accepted: 03/20/2015] [Indexed: 12/28/2022]
Abstract
Plasma protein binding (PPB) strongly affects drug distribution and pharmacokinetic behavior with consequences in overall pharmacological action. Extended plasma protein binding may be associated with drug safety issues and several adverse effects, like low clearance, low brain penetration, drug-drug interactions, loss of efficacy, while influencing the fate of enantiomers and diastereoisomers by stereoselective binding within the body. Therefore in holistic drug design approaches, where ADME(T) properties are considered in parallel with target affinity, considerable efforts are focused in early estimation of PPB mainly in regard to human serum albumin (HSA), which is the most abundant and most important plasma protein. The second critical serum protein α1-acid glycoprotein (AGP), although often underscored, plays also an important and complicated role in clinical therapy and thus the last years it has been studied thoroughly too. In the present review, after an overview of the principles of HSA and AGP binding as well as the structure topology of the proteins, the current trends and perspectives in the field of PPB predictions are presented and discussed considering both HSA and AGP binding. Since however for the latter protein systematic studies have started only the last years, the review focuses mainly to HSA. One part of the review highlights the challenge to develop rapid techniques for HSA and AGP binding simulation and their performance in assessment of PPB. The second part focuses on in silico approaches to predict HSA and AGP binding, analyzing and evaluating structure-based and ligand-based methods, as well as combination of both methods in the aim to exploit the different information and overcome the limitations of each individual approach. Ligand-based methods use the Quantitative Structure-Activity Relationships (QSAR) methodology to establish quantitate models for the prediction of binding constants from molecular descriptors, while they provide only indirect information on binding mechanism. Efforts for the establishment of global models, automated workflows and web-based platforms for PPB predictions are presented and discussed. Structure-based methods relying on the crystal structures of drug-protein complexes provide detailed information on the underlying mechanism but are usually restricted to specific compounds. They are useful to identify the specific binding site while they may be important in investigating drug-drug interactions, related to PPB. Moreover, chemometrics or structure-based modeling may be supported by experimental data a promising integrated alternative strategy for ADME(T) properties optimization. In the case of PPB the use of molecular modeling combined with bioanalytical techniques is frequently used for the investigation of AGP binding.
Collapse
|
40
|
Colclough N, Wenlock MC. Interpreting physicochemical experimental data sets. J Comput Aided Mol Des 2015; 29:779-94. [PMID: 26054297 DOI: 10.1007/s10822-015-9850-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/29/2015] [Indexed: 12/01/2022]
Abstract
With the wealth of experimental physicochemical data available to chemoinformaticians from the literature, commercial, and company databases an increasing challenge is the interpretation of such datasets. Subtle differences in experimental methodology used to generate these datasets can give rise to variations in physicochemical property values. Such methodology nuances will be apparent to an expert experimentalist but not necessarily to the data analyst and modeller. This paper describes the differences between common methodologies for measuring the four most important physicochemical properties namely aqueous solubility, octan-1-ol/water distribution coefficient, pK(a) and plasma protein binding highlighting key factors that can lead to systematic differences. Insight is given into how to identify datasets suitable for combining.
Collapse
Affiliation(s)
- Nicola Colclough
- Oncology and Drug Safety and Metabolism, Innovative Medicines, Mereside, AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.
| | - Mark C Wenlock
- Oncology and Drug Safety and Metabolism, Innovative Medicines, Mereside, AstraZeneca, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| |
Collapse
|
41
|
Walkup GK, You Z, Ross PL, Allen EKH, Daryaee F, Hale MR, O'Donnell J, Ehmann DE, Schuck VJA, Buurman ET, Choy AL, Hajec L, Murphy-Benenato K, Marone V, Patey SA, Grosser LA, Johnstone M, Walker SG, Tonge PJ, Fisher SL. Translating slow-binding inhibition kinetics into cellular and in vivo effects. Nat Chem Biol 2015; 11:416-23. [PMID: 25894085 DOI: 10.1038/nchembio.1796] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 03/19/2015] [Indexed: 01/18/2023]
Abstract
Many drug candidates fail in clinical trials owing to a lack of efficacy from limited target engagement or an insufficient therapeutic index. Minimizing off-target effects while retaining the desired pharmacodynamic (PD) response can be achieved by reduced exposure for drugs that display kinetic selectivity in which the drug-target complex has a longer half-life than off-target-drug complexes. However, though slow-binding inhibition kinetics are a key feature of many marketed drugs, prospective tools that integrate drug-target residence time into predictions of drug efficacy are lacking, hindering the integration of drug-target kinetics into the drug discovery cascade. Here we describe a mechanistic PD model that includes drug-target kinetic parameters, including the on- and off-rates for the formation and breakdown of the drug-target complex. We demonstrate the utility of this model by using it to predict dose response curves for inhibitors of the LpxC enzyme from Pseudomonas aeruginosa in an animal model of infection.
Collapse
Affiliation(s)
- Grant K Walkup
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Zhiping You
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Philip L Ross
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Eleanor K H Allen
- Institute for Chemical Biology and Drug Discovery, Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Fereidoon Daryaee
- Institute for Chemical Biology and Drug Discovery, Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Michael R Hale
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - John O'Donnell
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - David E Ehmann
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Virna J A Schuck
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Ed T Buurman
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Allison L Choy
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Laurel Hajec
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Kerry Murphy-Benenato
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Valerie Marone
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Sara A Patey
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Lena A Grosser
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Michele Johnstone
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| | - Stephen G Walker
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, New York, USA
| | - Peter J Tonge
- Institute for Chemical Biology and Drug Discovery, Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Stewart L Fisher
- Infection Innovative Medicines Unit, AstraZeneca Research and Development, Waltham, Massachusetts, USA
| |
Collapse
|
42
|
Patel YT, Jacus MO, Boulos N, Dapper JD, Davis AD, Vuppala PK, Freeman BB, Mohankumar KM, Throm SL, Gilbertson RJ, Stewart CF. Preclinical examination of clofarabine in pediatric ependymoma: intratumoral concentrations insufficient to warrant further study. Cancer Chemother Pharmacol 2015; 75:897-906. [PMID: 25724157 DOI: 10.1007/s00280-015-2713-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/19/2015] [Indexed: 01/16/2023]
Abstract
Clofarabine, a deoxyadenosine analog, was an active anticancer drug in our in vitro high-throughput screening against mouse ependymoma neurospheres. To characterize the clofarabine disposition in mice for further preclinical efficacy studies, we evaluated the plasma and central nervous system disposition in a mouse model of ependymoma. A plasma pharmacokinetic study of clofarabine (45 mg/kg, IP) was performed in CD1 nude mice bearing ependymoma to obtain initial plasma pharmacokinetic parameters. These estimates were used to derive D-optimal plasma sampling time points for cerebral microdialysis studies. A simulation of clofarabine pharmacokinetics in mice and pediatric patients suggested that a dosage of 30 mg/kg IP in mice would give exposures comparable to that in children at a dosage of 148 mg/m(2). Cerebral microdialysis was performed to study the tumor extracellular fluid (ECF) disposition of clofarabine (30 mg/kg, IP) in the ependymoma cortical allografts. Plasma and tumor ECF concentration-time data were analyzed using a nonlinear mixed effects modeling approach. The median unbound fraction of clofarabine in mouse plasma was 0.79. The unbound tumor to plasma partition coefficient (K pt,uu: ratio of tumor to plasma AUCu,0-inf) of clofarabine was 0.12 ± 0.05. The model-predicted mean tumor ECF clofarabine concentrations were below the in vitro 1-h IC50 (407 ng/mL) for ependymoma neurospheres. Thus, our results show the clofarabine exposure reached in the tumor ECF was below that associated with an antitumor effect in our in vitro washout study. Therefore, clofarabine was de-prioritized as an agent to treat ependymoma, and further preclinical studies were not pursued.
Collapse
Affiliation(s)
- Yogesh T Patel
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tedesco D, Bertucci C. Induced circular dichroism as a tool to investigate the binding of drugs to carrier proteins: Classic approaches and new trends. J Pharm Biomed Anal 2015; 113:34-42. [PMID: 25769668 DOI: 10.1016/j.jpba.2015.02.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 10/24/2022]
Abstract
Induced circular dichroism (ICD) is a spectroscopic phenomenon that provides versatile and useful methods for characterizing the structural and dynamic properties of the binding of drugs to target proteins. The understanding of biorecognition processes at the molecular level is essential to discover and validate new pharmacological targets, and to design and develop new potent and selective drugs. The present article reviews the main applications of ICD to drug binding studies on serum carrier proteins, going from the classic approaches for the derivation of drug binding parameters and the identification of binding sites, to an overview of the emerging trends for the characterization of binding modes by means of quantum chemical (QC) techniques. The advantages and limits of the ICD methods for the determination of binding parameters are critically reviewed; the capability to investigate the binding interactions of drugs and metabolites to their target proteins is also underlined, as well as the possibility of characterizing the binding sites to obtain a complete picture of the binding mechanism and dynamics. The new applications of ICD methods to identify stereoselective binding modes of drug/protein complexes are then reviewed with relevant examples. The combined application of experimental ICD spectroscopy and QC calculations is shown to identify qualitatively the bound conformations of ligands to target proteins even in the absence of a detailed structure of the binding sites, either obtained from experimental X-ray crystallography and NMR measurements or from computational models of the complex.
Collapse
Affiliation(s)
- Daniele Tedesco
- Department of Pharmacy and Biotechnology, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy
| | - Carlo Bertucci
- Department of Pharmacy and Biotechnology, University of Bologna, via Belmeloro 6, 40126 Bologna, Italy.
| |
Collapse
|
44
|
Metabolic characterization of meso-dihydroguaiaretic acid in liver microsomes and in mice. Food Chem Toxicol 2015; 76:94-102. [DOI: 10.1016/j.fct.2014.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 11/17/2022]
|
45
|
Ritchie HE, Svensson CH, Nilsson MF, Webster WS. A comparison of drug-induced cardiotoxicity in rat embryos cultured in human serum or protein free media. J Pharmacol Toxicol Methods 2014; 70:276-82. [DOI: 10.1016/j.vascn.2014.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/07/2014] [Accepted: 07/23/2014] [Indexed: 10/25/2022]
|
46
|
Liu X, Wright M, Hop CECA. Rational use of plasma protein and tissue binding data in drug design. J Med Chem 2014; 57:8238-48. [PMID: 25099658 DOI: 10.1021/jm5007935] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
It is a commonly accepted assumption that only unbound drug molecules are available to interact with their targets. Therefore, one of the objectives in drug design is to optimize the compound structure to increase in vivo unbound drug concentration. In this review, theoretical analyses and experimental observations are presented to illustrate that low plasma protein binding does not necessarily lead to high in vivo unbound plasma concentration. Similarly, low brain tissue binding does not lead to high in vivo unbound brain tissue concentration. Instead, low intrinsic clearance leads to high in vivo unbound plasma concentration, and low efflux transport activity at the blood-brain barrier leads to high unbound brain concentration. Plasma protein and brain tissue binding are very important parameters in understanding pharmacokinetics, pharmacodynamics, and toxicities of drugs, but these parameters should not be targeted for optimization in drug design.
Collapse
Affiliation(s)
- Xingrong Liu
- Genentech, Inc. , South San Francisco, California 94080, United States
| | | | | |
Collapse
|
47
|
Ossipov K, Scaffidi-Domianello YY, Seregina IF, Galanski MS, Keppler BK, Timerbaev AR, Bolshov MA. Inductively coupled plasma mass spectrometry for metallodrug development: albumin binding and serum distribution of cytotoxic cis- and trans-isomeric platinum(II) complexes. J Inorg Biochem 2014; 137:40-5. [PMID: 24803025 DOI: 10.1016/j.jinorgbio.2014.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/11/2014] [Accepted: 04/11/2014] [Indexed: 01/31/2023]
Abstract
Binding to plasma proteins is one of the major metabolic pathways of metallodrugs. In the case of platinum-based anticancer drugs, it is the interaction with serum albumin that affects most strongly their in vivo behavior. Since both the configuration, i.e. cis-trans-isomerism, and the nature of leaving groups have an effect on the reactivity of Pt(II) coordination compounds toward biomolecules, a set of cis- and trans-configured complexes with halide leaving groups (Cl(-), Br(-), and I(-)) and 2-propanone oxime as carrier ligands was chosen for this study. Binding experiments were performed both with albumin and human serum and the Pt content in ultrafiltrates was quantified using inductively coupled plasma mass spectrometry. In order to shed light on the binding mechanism, the albumin binding constant (KHSA) and the octanol-water partition coefficient (P) were experimentally determined and relationships between log KHSA and log P were explored. The correlation was found significant only for cis-configured platinum complexes (R(2)=0.997 and standard deviation=0.02), indicating a certain contribution of the nonspecific binding which is largely dominated by the lipophilicity of compounds. In contrast, for trans-complexes a specific molecular recognition element plays a significant role. The participation of albumin in drug distribution in blood serum was assessed using an equilibrium distribution model and by comparing the percentage binding in the albumin and serum-protein fractions. Irrespective of the compound polarity, albumin contributes from 85 to 100% to the overall binding in serum.
Collapse
Affiliation(s)
- Konstantin Ossipov
- Division of Analytical Chemistry, Chemistry Department, Moscow State University, Leninskie Gory 1, 119992 Moscow, Russia
| | | | - Irina F Seregina
- Division of Analytical Chemistry, Chemistry Department, Moscow State University, Leninskie Gory 1, 119992 Moscow, Russia
| | - Mathea S Galanski
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
| | - Andrei R Timerbaev
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria; Vernadsky Institute of Geochemistry and Analytical Chemistry, Russian Academy of Sciences, Kosygin Str. 19, 119991 Moscow, Russia.
| | - Mikhail A Bolshov
- Division of Analytical Chemistry, Chemistry Department, Moscow State University, Leninskie Gory 1, 119992 Moscow, Russia; Institute for Spectroscopy, Russian Academy of Sciences, Fizicheskaya 5, 142190 Troitsk (Moscow Region), Russia
| |
Collapse
|
48
|
Borijihan G, Li Y, Gao J, Bao JJ. Development of a novel 96-well format for liquid-liquid microextraction and its application in the HPLC analysis of biological samples. J Sep Sci 2014; 37:1155-61. [DOI: 10.1002/jssc.201300954] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/14/2014] [Accepted: 02/16/2014] [Indexed: 11/05/2022]
Affiliation(s)
- Guirong Borijihan
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering; School of Pharmaceutical Science and Technology; Tianjin University; Tianjin China
| | - Youxin Li
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering; School of Pharmaceutical Science and Technology; Tianjin University; Tianjin China
| | - Jianguo Gao
- Qingdao Entry-Exit Inspection Quarantine Bureau; Shandong China
| | - James J. Bao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, Collaborative Innovation Center of Chemical Science and Engineering; School of Pharmaceutical Science and Technology; Tianjin University; Tianjin China
| |
Collapse
|
49
|
Gasymov OK, Abduragimov AR, Glasgow BJ. A simple model-free method for direct assessment of fluorescent ligand binding by linear spectral summation. J Fluoresc 2013; 24:231-8. [PMID: 24043458 DOI: 10.1007/s10895-013-1290-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
Abstract
Fluorescent tagged ligands are commonly used to determine binding to proteins. However, bound and free ligand concentrations are not directly determined. Instead the response in a fluorescent ligand titration experiment is considered to be proportional to the extent of binding and, therefore, the maximum value of binding is scaled to the total protein concentration. Here, a simple model-free method is presented to be performed in two steps. In the first step, normalized bound and free spectra of the ligand are determined. In the second step, these spectra are used to fit composite spectra as the sum of individual components or linear spectral summation. Using linear spectral summation, free and bound 1-Anilinonaphthalene-8-Sulfonic Acid (ANS) fluorescent ligand concentrations are directly calculated to determine ANS binding to tear lipocalin (TL), an archetypical ligand binding protein. Error analysis shows that the parameters that determine bound and free ligand concentrations were recovered with high certainty. The linear spectral summation method is feasible when fluorescence intensity is accompanied by a spectral shift upon protein binding. Computer simulations of the experiments of ANS binding to TL indicate that the method is feasible when the fluorescence spectral shift between bound and free forms of the ligand is just 8 nm. Ligands tagged with environmentally sensitive fluorescent dyes, e.g., dansyl chromophore, are particularly suitable for this method.
Collapse
Affiliation(s)
- Oktay K Gasymov
- Departments of Ophthalmology, Pathology and Laboratory Medicine, Jules Stein Eye Institute, University of California at Los Angeles, Los Angeles, CA, 90095, USA,
| | | | | |
Collapse
|
50
|
Filip Z, Jan K, Vendula S, Jana KZ, Kamil M, Kamil K. Albumin and α1-acid glycoprotein: old acquaintances. Expert Opin Drug Metab Toxicol 2013; 9:943-54. [DOI: 10.1517/17425255.2013.790364] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|