1
|
Activation of the GLP-1 receptor by chloropyrimidine derivatives. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
2
|
AlNeyadi SS, Amir N, Ghattas MA, Atatreh N, Alketbi SS, Ajeil RA, Adem A. Controlled Release of Pyrimidine Compound Using Polymeric Coated ZIF-8 Metal-Organic Framework as Glucagon-Like Peptide-1 Receptor Agonist Carrier. Molecules 2020; 25:E4313. [PMID: 32962260 PMCID: PMC7570959 DOI: 10.3390/molecules25184313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022] Open
Abstract
This work demonstrates synthetic strategies for the incorporation of a synthesized pyrimidine glucagon-like peptide-1 (GLP-1) agonist into alginate-coated ZIF-8. The prepared pyrimidine GLP-1 agonist used for the treatment of diabetes type II, was trapped inside polymer coated ZIF-8. The encapsulation of the GLP-1 agonist was confirmed by UV-visible and FT-IR spectroscopies. Furthermore, the release kinetics of GLP-1 agonist drug from alginate-coated ZIF-8 were investigated in phosphate-buffered saline at 37 °C at pH 8 and 1.5. The alginate-coated ZIF-8 exhibited much faster drug release at basic pH than at pH 1.5, indicating the potential of the alginate-coated ZIF-8 system to overcome the fast degradation at acidic pH of the stomach and improve the drug's activity. This study may open the way for the synthesis of new metal organic frameworks (MOFs) to enhance drug delivery systems.
Collapse
Affiliation(s)
- Shaikha S. AlNeyadi
- Department of Chemistry, College of Science, UAE University, Al-Ain P.O. Box 15551, UAE; (S.S.A.); (R.A.A.)
| | - Naheed Amir
- Department of Pharmacology, College of Health and Science, UAE University, Al-Ain P.O. Box 17666, UAE; (N.A.); (A.A.)
| | - Mohammad A. Ghattas
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, UAE; (M.A.G.); (N.A.)
| | - Noor Atatreh
- College of Pharmacy, Al Ain University, Abu Dhabi P.O. Box 112612, UAE; (M.A.G.); (N.A.)
| | - Shaikha S. Alketbi
- Department of Chemistry, College of Science, UAE University, Al-Ain P.O. Box 15551, UAE; (S.S.A.); (R.A.A.)
| | - Ruba Al Ajeil
- Department of Chemistry, College of Science, UAE University, Al-Ain P.O. Box 15551, UAE; (S.S.A.); (R.A.A.)
| | - Abdu Adem
- Department of Pharmacology, College of Health and Science, UAE University, Al-Ain P.O. Box 17666, UAE; (N.A.); (A.A.)
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, UAE
| |
Collapse
|
3
|
Chong Y, Chang J, Zhao W, He Y, Li Y, Zhang H, Qi C. Synthesis and evaluation of novel 18 F-labeled quinazoline derivatives with low lipophilicity for tumor PET imaging. J Labelled Comp Radiopharm 2018; 61:42-53. [PMID: 28833405 DOI: 10.1002/jlcr.3538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/14/2017] [Accepted: 07/31/2017] [Indexed: 01/06/2023]
Abstract
Four novel 18 F-labeled quinazoline derivatives with low lipophilicity, [18 F]4-(2-fluoroethoxy)-6,7-dimethoxyquinazoline ([18 F]I), [18 F]4-(3-((4-(2-fluoroethoxy)-7-methoxyquinazolin-6-yl)oxy)propyl)morpholine ([18 F]II), [18 F]4-(2-fluoroethoxy)-7-methoxy-6-(2-methoxyethoxy)quinazoline ([18 F]III), and [18 F]4-(2-fluoroethoxy)-6,7-bis(2-methoxyethoxy)quinazoline ([18 F]IV), were synthesized via a 2-step radiosynthesis procedure with an overall radiochemical yield of 10% to 38% (without decay correction) and radiochemical purities of >98%. The lipophilicity and stability of labeled compounds were tested in vitro. The log P values of the 4 radiotracers ranged from 0.52 to 1.07. We then performed ELISA to measure their affinities to EGFR-TK; ELISA assay results indicated that each inhibitor was specifically bounded to EGFR-TK in a dose-dependent manner. The EGFR-TK autophosphorylation IC50 values of [18 F]I, [18 F]II, [18 F]III, and [18 F]IV were 7.732, 0.4698, 0.1174, and 0.1176 μM, respectively. All labeled compounds were evaluated via cellular uptake and blocking studies in HepG2 cell lines in vitro. Cellular uptake and blocking experiment results indicated that [18 F]I and [18 F]III had excellent cellular uptake at 120-minute postinjection in HepG2 carcinoma cells (51.80 ± 3.42%ID/mg protein and 27.31 ± 1.94%ID/mg protein, respectively). Additionally, biodistribution experiments in S180 tumor-bearing mice in vivo indicated that [18 F]I had a very fast clearance in blood and a relatively high uptake ratio of tumor to blood (4.76) and tumor to muscle (1.82) at 60-minute postinjection. [18 F]III had a quick clearance in plasma, and its highest uptake ratio of tumor to muscle was 2.55 at 15-minute postinjection. These experimental results and experiences were valuable for the further exploration of novel radiotracers of quinazoline derivatives.
Collapse
Affiliation(s)
- Yan Chong
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Jin Chang
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Wenwen Zhao
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Yong He
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Yuqiao Li
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Huabei Zhang
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| | - Chuanmin Qi
- Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, People's Republic of China
| |
Collapse
|
4
|
Synthesis and evaluation of radiolabeled AGI-5198 analogues as candidate radiotracers for imaging mutant IDH1 expression in tumors. Bioorg Med Chem Lett 2018; 28:694-699. [PMID: 29366652 DOI: 10.1016/j.bmcl.2018.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 11/23/2022]
Abstract
Mutations in the metabolic enzyme isocitrate dehydrogenase 1 (IDH1) are commonly found in gliomas. AGI-5198, a potent and selective inhibitor of the mutant IDH1 enzyme, was radiolabeled with radioiodine and fluorine-18. These radiotracers were evaluated as potential probes for imaging mutant IDH1 expression in tumors with positron emission tomography (PET). Radioiodination of AGI-5198 was achieved using a tin precursor in 79 ± 6% yield (n = 9), and 18F-labeling was accomplished by the Ugi reaction in a decay-corrected radiochemical yield of 2.6 ± 1.6% (n = 5). The inhibitory potency of the analogous nonradioactive compounds against mutant IDH1 (IDH1-R132H) was determined in enzymatic assays. Cell uptake studies using radiolabeled AGI-5198 analogues revealed somewhat higher uptake in IDH1-mutated cells than that in wild-type IDH1 cells. The radiolabeled compounds displayed favorable tissue distribution characteristics in vivo, and good initial uptake in IDH1-mutated tumor xenografts; however, tumor uptake decreased with time. Radioiodinated AGI-5198 exhibited higher tumor-to-background ratios compared with 18F-labeled AGI-5198; unfortunately, similar results were observed in wild-type IDH1 tumor xenografts as well, indicating lack of selectivity for mutant IDH1 for this tracer. These results suggest that AGI-5198 analogues are not a promising platform for radiotracer development. Nonetheless, insights gained from this study may help in design and optimization of novel chemical scaffolds for developing radiotracers for imaging the mutant IDH1 enzyme.
Collapse
|
5
|
AlNeyadi SS, Salem AA, Ghattas MA, Atatreh N, Abdou IM. Antibacterial activity and mechanism of action of the benzazole acrylonitrile-based compounds: In vitro , spectroscopic, and docking studies. Eur J Med Chem 2017; 136:270-282. [DOI: 10.1016/j.ejmech.2017.05.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/18/2023]
|
6
|
van der Born D, Pees A, Poot AJ, Orru RVA, Windhorst AD, Vugts DJ. Fluorine-18 labelled building blocks for PET tracer synthesis. Chem Soc Rev 2017; 46:4709-4773. [DOI: 10.1039/c6cs00492j] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents a comprehensive overview of the synthesis and application of fluorine-18 labelled building blocks since 2010.
Collapse
Affiliation(s)
- Dion van der Born
- Department of Radiology & Nuclear Medicine
- VU University Medical Center
- 1081 HV Amsterdam
- The Netherlands
| | - Anna Pees
- Department of Radiology & Nuclear Medicine
- VU University Medical Center
- 1081 HV Amsterdam
- The Netherlands
| | - Alex J. Poot
- Department of Radiology & Nuclear Medicine
- VU University Medical Center
- 1081 HV Amsterdam
- The Netherlands
| | - Romano V. A. Orru
- Department of Chemistry and Pharmaceutical Sciences and Amsterdam Institute for Molecules
- Medicines & Systems (AIMMS)
- VU University Amsterdam
- Amsterdam
- The Netherlands
| | - Albert D. Windhorst
- Department of Radiology & Nuclear Medicine
- VU University Medical Center
- 1081 HV Amsterdam
- The Netherlands
| | - Danielle J. Vugts
- Department of Radiology & Nuclear Medicine
- VU University Medical Center
- 1081 HV Amsterdam
- The Netherlands
| |
Collapse
|
7
|
Recent Advances in the Development and Application of Radiolabeled Kinase Inhibitors for PET Imaging. Molecules 2015; 20:22000-27. [PMID: 26690113 PMCID: PMC6332294 DOI: 10.3390/molecules201219816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Over the last 20 years, intensive investigation and multiple clinical successes targeting protein kinases, mostly for cancer treatment, have identified small molecule kinase inhibitors as a prominent therapeutic class. In the course of those investigations, radiolabeled kinase inhibitors for positron emission tomography (PET) imaging have been synthesized and evaluated as diagnostic imaging probes for cancer characterization. Given that inhibitor coverage of the kinome is continuously expanding, in vivo PET imaging will likely find increasing applications for therapy monitoring and receptor density studies both in- and outside of oncological conditions. Early investigated radiolabeled inhibitors, which are mostly based on clinically approved tyrosine kinase inhibitor (TKI) isotopologues, have now entered clinical trials. Novel radioligands for cancer and PET neuroimaging originating from novel but relevant target kinases are currently being explored in preclinical studies. This article reviews the literature involving radiotracer design, radiochemistry approaches, biological tracer evaluation and nuclear imaging results of radiolabeled kinase inhibitors for PET reported between 2010 and mid-2015. Aspects regarding the usefulness of pursuing selective vs. promiscuous inhibitor scaffolds and the inherent challenges associated with intracellular enzyme imaging will be discussed.
Collapse
|
8
|
Abstract
The logic of total synthesis transformed a stagnant state of medicinal and synthetic organic chemistry when there was a paucity of methods and reagents to synthesize drug molecules and/or natural products. Molecular imaging by positron emission tomography (PET) is now experiencing a renaissance in the way radiopharmaceuticals for molecular imaging are synthesized, however, a paradigm shift is desperately needed in the discovery pipeline to accelerate in vivo imaging studies. A significant challenge in radiochemistry is the limited choice of labeled reagents (or building blocks) available for the synthesis of novel radiopharmaceuticals with the most commonly used short-lived radionuclides carbon-11 (11C; half-life ~20 minutes) and fluorine-18 (18F; half-life ~2 hours). In fact, most drugs cannot be labeled with 11C or 18F due to a lack of efficient and diverse radiosynthetic methods. In general, routine radiopharmaceutical production relies on the incorporation of the isotope at the last or penultimate step of synthesis, ideally within one half-life of the radionuclide, to maximize radiochemical yields and specific activities thereby reducing losses due to radioactive decay. Reliance on radiochemistry conducted within the constraints of an automated synthesis unit ("box") has stifled the exploration of multi-step reactions with short-lived radionuclides. Radiopharmaceutical synthesis can be transformed by considering logic of total synthesis to develop novel approaches for 11C- and 18F-radiolabeling complex molecules via retrosynthetic analysis and multi-step reactions. As a result of such exploration, new methods, reagents and radiopharmaceuticals for in vivo imaging studies are discovered. A new avenue to develop radiotracers that were previously unattainable due to the lack of efficient radiosynthetic methods is necessary to work towards our ultimate, albeit impossible goal - the concept we term total radiosynthesis - to radiolabel virtually any molecule. As with the vast majority of drugs, most radiotracers also fail, therefore expeditious evaluation of tracers in preclinical models prior to optimization or derivatization of the lead molecules/drugs is necessary. Furthermore the exact position of the 11C and 18F radionuclide in tracers is often critical for metabolic considerations, and flexible methodologies to introduce the radiolabel are needed. Using the principles of total synthesis our laboratory and others have shown that multi-step radiochemical reactions are indeed suitable for preclinical and even clinical use. As the goal of total synthesis is to be concise, we have also simplified the syntheses of radiopharmaceuticals. We are presently developing new strategies via [11C]CO2 fixation which has enabled library radiosynthesis as well as labeling non-activated arenes using [18F]fluoride via iodonium ylides. Both of which have proven to be suitable for human PET imaging. We concurrently utilize state-of-the-art automation technologies including microfluidic flow chemistry and rapid purification strategies for radiopharmaceutical production. In this account we highlight how total radiosynthesis has impacted our radiochemistry program, with prominent examples from others, focusing on its impact towards preclinical and clinical research studies.
Collapse
Affiliation(s)
- Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit St., Boston, MA, 02114, USA
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit St., Boston, MA, 02114, USA
| |
Collapse
|
9
|
Lau J, Liu Z, Lin KS, Pan J, Zhang Z, Vullo D, Supuran CT, Perrin DM, Bénard F. Trimeric Radiofluorinated Sulfonamide Derivatives to Achieve In Vivo Selectivity for Carbonic Anhydrase IX–Targeted PET Imaging. J Nucl Med 2015. [DOI: 10.2967/jnumed.114.153288] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
10
|
Quantitative analysis of [11C]-erlotinib PET demonstrates specific binding for activating mutations of the EGFR kinase domain. Neoplasia 2014; 15:1347-53. [PMID: 24403856 DOI: 10.1593/neo.131666] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/07/2013] [Accepted: 11/10/2013] [Indexed: 11/18/2022] Open
Abstract
Activating mutations of the epidermal growth factor receptor (EGFR) occur in multiple tumor types, including non-small cell lung cancer (NSCLC) and malignant glioma, and have become targets for therapeutic intervention. The determination of EGFR mutation status using a noninvasive, molecular imaging approach has the potential for clinical utility. In this study, we investigated [(11)C]-erlotinib positron emission tomography (PET) imaging as a tool to identify activating mutations of EGFR in both glioma and NSCLC xenografts. Radiotracer specific binding was determined for high and low specific activity (SA) [(11)C]-erlotinib PET scans in mice bearing synchronous human cancer xenografts with different EGFR expression profiles (PC9, HCC827, U87, U87 ΔEGFR, and SW620). Although xenograft immunohistochemistry demonstrated constitutive EGFR phosphorylation, PET scan analysis using the Simplified Reference Tissue Model showed that only kinase domain mutant NSCLC (HCC827 and PC9) had significantly greater binding potentials in high versus low SA scans. Xenografts with undetectable EGFR expression (SW620), possessing wild-type EGFR (U87), and expressing an activating extracellular domain mutation (U87 ΔEGFR) were indistinguishable under both high and low SA scan conditions. The results suggest that [(11)C]-erlotinib is a promising radiotracer that could provide a novel clinical methodology for assessing EGFR and erlotinib interactions in patients with tumors that harbor EGFR-activating kinase domain mutations.
Collapse
|
11
|
Rotstein BH, Stephenson NA, Vasdev N, Liang SH. Spirocyclic hypervalent iodine(III)-mediated radiofluorination of non-activated and hindered aromatics. Nat Commun 2014; 5:4365. [PMID: 25007318 DOI: 10.1038/ncomms5365] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/10/2014] [Indexed: 01/21/2023] Open
Abstract
Fluorine-18 (t½=109.7 min) is the most commonly used isotope to prepare radiopharmaceuticals for molecular imaging by positron emission tomography (PET). Nucleophilic aromatic substitution reactions of suitably activated (electron-deficient) aromatic substrates with no-carrier-added [(18)F]fluoride ion are routinely carried out in the synthesis of radiotracers in high specific activities. Despite extensive efforts to develop a general (18)F-labelling technique for non-activated arenes there is an urgent and unmet need to achieve this goal. Here we describe an effective solution that relies on the chemistry of spirocyclic hypervalent iodine(III) complexes, which serve as precursors for rapid, one-step regioselective radiofluorination with [(18)F]fluoride. This methodology proves to be efficient for radiolabelling a diverse range of non-activated functionalized arenes and heteroarenes, including arene substrates bearing electron-donating groups, bulky ortho functionalities, benzylic substituents and meta-substituted electron-withdrawing groups. Polyfunctional molecules and a range of previously elusive (18)F-labelled building blocks, compounds and radiopharmaceuticals are synthesized.
Collapse
Affiliation(s)
- Benjamin H Rotstein
- 1] Division of Nuclear Medicine and Molecular Imaging, Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA [2]
| | - Nickeisha A Stephenson
- 1] Division of Nuclear Medicine and Molecular Imaging, Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA [2]
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Center for Advanced Medical Imaging Sciences, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
12
|
Liu L, Jalili N, Baergen A, Ng S, Bailey J, Derda R, Klassen JS. Fluorine bonding enhances the energetics of protein-lipid binding in the gas phase. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2014; 25:751-757. [PMID: 24658801 DOI: 10.1007/s13361-014-0837-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 06/03/2023]
Abstract
This paper reports on the first experimental study of the energies of noncovalent fluorine bonding in a protein-ligand complex in the absence of solvent. Arrhenius parameters were measured for the dissociation of gaseous deprotonated ions of complexes of bovine β-lactoglobulin (Lg), a model lipid-binding protein, and four fluorinated analogs of stearic acid (SA), which contained (X =) 13, 15, 17, or 21 fluorine atoms. In all cases, the activation energies (E(a)) measured for the loss of neutral XF-SA from the (Lg + XF-SA)⁷⁻ ions are larger than for SA. From the kinetic data, the average contribution of each > CF₂ group to E(a) was found to be ~1.1 kcal mol⁻¹, which is larger than the ~0.8 kcal mol⁻¹ value reported for > CH₂ groups. Based on these results, it is proposed that fluorocarbon–protein interactions are inherently stronger (enthalpically) than the corresponding hydrocarbon interactions.
Collapse
|
13
|
|
14
|
Liang SH, Collier TL, Rotstein BH, Lewis R, Steck M, Vasdev N. Rapid microfluidic flow hydrogenation for reduction or deprotection of 18F-labeled compounds. Chem Commun (Camb) 2014; 49:8755-7. [PMID: 23948863 DOI: 10.1039/c3cc45166f] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We have combined the benefits of both microfluidics and flow hydrogenation to provide facile access to previously underutilized reduction and protecting group chemistries for PET imaging applications. The rapid removal of an O-benzyl protecting group to prepare 2-[(18)F]fluoroquinolin-8-ol and the reduction of a nitro group in the synthesis of 4-[(18)F]fluoroaniline were achieved within 3 minutes.
Collapse
Affiliation(s)
- Steven H Liang
- Department of Radiology, Harvard Medical School, 55 Fruit Street, Boston, MA, USA 02114.
| | | | | | | | | | | |
Collapse
|
15
|
Coecke S, Pelkonen O, Leite SB, Bernauer U, Bessems JG, Bois FY, Gundert-Remy U, Loizou G, Testai E, Zaldívar JM. Toxicokinetics as a key to the integrated toxicity risk assessment based primarily on non-animal approaches. Toxicol In Vitro 2012; 27:1570-7. [PMID: 22771339 DOI: 10.1016/j.tiv.2012.06.012] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 03/09/2012] [Accepted: 06/22/2012] [Indexed: 02/02/2023]
Abstract
Toxicokinetics (TK) is the endpoint that informs about the penetration into and fate within the body of a toxic substance, including the possible emergence of metabolites. Traditionally, the data needed to understand those phenomena have been obtained in vivo. Currently, with a drive towards non-animal testing approaches, TK has been identified as a key element to integrate the results from in silico, in vitro and already available in vivo studies. TK is needed to estimate the range of target organ doses that can be expected from realistic human external exposure scenarios. This information is crucial for determining the dose/concentration range that should be used for in vitro testing. Vice versa, TK is necessary to convert the in vitro results, generated at tissue/cell or sub-cellular level, into dose response or potency information relating to the entire target organism, i.e. the human body (in vitro-in vivo extrapolation, IVIVE). Physiologically based toxicokinetic modelling (PBTK) is currently regarded as the most adequate approach to simulate human TK and extrapolate between in vitro and in vivo contexts. The fact that PBTK models are mechanism-based which allows them to be 'generic' to a certain extent (various extrapolations possible) has been critical for their success so far. The need for high-quality in vitro and in silico data on absorption, distribution, metabolism as well as excretion (ADME) as input for PBTK models to predict human dose-response curves is currently a bottleneck for integrative risk assessment.
Collapse
Affiliation(s)
- Sandra Coecke
- ECVAM, Institute for Health & Consumer Protection, European Commission Joint Research Centre, 21027 Ispra (VA), Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Vasdev N, Cao P, van Oosten EM, Wilson AA, Houle S, Hao G, Sun X, Slavine N, Alhasan M, Antich PP, Bonte FJ, Kulkarni P. Synthesis and PET imaging studies of [18F]2-fluoroquinolin-8-ol ([18F]CABS13) in transgenic mouse models of Alzheimer's disease. MEDCHEMCOMM 2012. [DOI: 10.1039/c2md20075a] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|