Generic placeholder image

Current Chemical Biology

Editor-in-Chief

ISSN (Print): 2212-7968
ISSN (Online): 1872-3136

Research Article

Dose Formulation, Biodistribution and PET Imaging Studies of a First-in-Class Fluorine-18 Organophosphorus Cholinesterase Inhibitor Tracer in Rat

Author(s): Kiel D. Neumann, Joseph E. Blecha, Chih-Kai Chao, Tony Huynh, Kurt R. Zinn, Henry F. VanBrocklin, Charles M. Thompson and John M. Gerdes*

Volume 14, Issue 4, 2020

Page: [289 - 303] Pages: 15

DOI: 10.2174/2212796814999201005195509

open access plus

Abstract

Background: To investigate dynamic live tissue organophosphorus nerve agent uptake and distribution fates resulting in acetylcholinesterase inhibition, we recently reported the first-in-class fluorine-18 [18F] radiolabeled Positron Emission Tomography (PET) imaging tracer known as [18F]O-(2-fluoroethyl)-O-(p-nitrophenyl)methylphosphonate. This tracer has been initially studied in live rats with PET imaging.

Objective: We sought to evaluate the PET tracer in vivo using a new dose formulation of saline, ethanol and L-ascorbic acid, and compare the influence of this formulation on in vivo tracer performance to previous data collected using a CH3CN:PBS formulation.

Methods: A high molar activity [18F]tracer radiosynthesis was used. Doses were formulated as saline, ethanol (≤ 1%) and L-ascorbic acid (0.1%), pH 4.0-4.5. Stability was evaluated to 6 h. Dose injection (i.v.) into male rats was followed by either ex vivo biodistribution profiling at 5, 30, 90 min, or dynamic 90 min PET imaging. Rat biodistribution and PET imaging data were compared.

Results and Discussion: An optimized radiosynthesis (8 ± 2 % RCY) resulted in stable doses for 6 h (>99%). Arterial blood included a tracer and a single metabolite. The ex vivo biodistribution and live tissue PET imaging data revealed rapid radioactivity uptake and distributed tissue levels: heart and lung, highest; liver, moderate; and brain, lowest.

Conclusion: Imaging and biodistribution data were highly correlated with expected radioactivity tissue uptake and distribution in target organs. Lower brain radioactivity levels by PET imaging were found for the new formulation (saline, 1% L-ascorbic acid, < 1% ethanol) as compared to the established CH3CN:PBS formulation. Overall, we found that the i.v. dose formulation changed the in vivo profile of an organophosphorus PET tracer that is considered an important finding for future organophosphorus PET tracer studies.

Keywords: Fluorine-18, organophosphorus, acetylcholinesterase, adduct, positron emission tomography (pet), biodistribution, L-ascorbic acid, rat.

Graphical Abstract
[1]
Casida JE, Quistad GB. Serine hydrolase targets of organophosphorus toxicants. Chem Biol Interact 2005; 157-158: 277-83.
[http://dx.doi.org/10.1016/j.cbi.2005.10.036] [PMID: 16243304]
[2]
Bajgar J. Organophosphates/nerve agent poisoning: mechanism of action, diagnosis, prophylaxis, and treatment. Adv Clin Chem 2004; 38: 151-216.
[http://dx.doi.org/10.1016/S0065-2423(04)38006-6] [PMID: 15521192]
[3]
Ballantyne B, Marrs TC. Clinical and experimental toxicology of organophosphates and carbamates. Boston, MA: Butterworth Heinemann 1992.
[4]
Colović MB, Krstić DZ, Lazarević-Pašti TD, Bondžić AM, Vasić VM. Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol 2013; 11(3): 315-35.
[http://dx.doi.org/10.2174/1570159X11311030006] [PMID: 24179466]
[5]
Coban A, Carr RL, Chambers HW, Willeford KO, Chambers JE. Comparison of inhibition kinetics of several organophosphates, including some nerve agent surrogates, using human erythrocyte and rat and mouse brain acetylcholinesterase. Toxicol Lett 2016; 248: 39-45.
[http://dx.doi.org/10.1016/j.toxlet.2016.03.002] [PMID: 26965078]
[6]
Elhanany E, Ordentlich A, Dgany O, et al. Resolving pathways of interaction of covalent inhibitors with the active site of acetylcholinesterases: MALDI-TOF/MS analysis of various nerve agent phosphyl adducts. Chem Res Toxicol 2001; 14(7): 912-8.
[http://dx.doi.org/10.1021/tx0100542] [PMID: 11453739]
[7]
Chao C-K, Balasubramanian N, Gerdes JM, Thompson CM. The inhibition, reactivation and mechanism of VX-, sarin-, fluoro-VX and fluoro-sarin surrogates following their interaction with HuAChE and HuBuChE. Chem Biol Interact 2018; 291: 220-7.
[http://dx.doi.org/10.1016/j.cbi.2018.06.019] [PMID: 29920286]
[8]
Chao C-K, Ahmed SK, Gerdes JM, Thompson CM. Novel organophosphate ligand O-(2-fluoroethyl)-O-(p-nitrophenyl)methylphosphonate: synthesis, hydrolytic stability and analysis of the inhibition and reactivation of cholinesterases. Chem Res Toxicol 2016; 29(11): 1810-7.
[http://dx.doi.org/10.1021/acs.chemrestox.6b00160] [PMID: 27551891]
[9]
Chambers JE, Meek EC, Chambers HW. Novel brain-penetrating oximes for reactivation of cholinesterase inhibited by sarin and VX surrogates. Ann N Y Acad Sci 2016; 1374(1): 52-8.
[http://dx.doi.org/10.1111/nyas.13053] [PMID: 27153507]
[10]
Chambers JE, Chambers HW, Meek EC, Pringle RB. Testing of novel brain-penetrating oxime reactivators of acetylcholinesterase inhibited by nerve agent surrogates. Chem Biol Interact 2013; 203(1): 135-8.
[http://dx.doi.org/10.1016/j.cbi.2012.10.017] [PMID: 23123249]
[11]
James SL, Ahmed SK, Murphy S, et al. A novel fluorine-18 β-fluoroethoxy organophosphate positron emission tomography imaging tracer targeted to central nervous system acetylcholinesterase. ACS Chem Neurosci 2014; 5(7): 519-24.
[http://dx.doi.org/10.1021/cn500024c] [PMID: 24716794]
[12]
Kaleem Ahmed S, Belabassi Y, Sankaranarayanan L, Chao CK, Gerdes JM, Thompson CM. Synthesis and anti-acetylcholinesterase properties of novel β- and γ-substituted alkoxy organophosphonates. Bioorg Med Chem Lett 2013; 23(7): 2048-51.
[http://dx.doi.org/10.1016/j.bmcl.2013.02.010] [PMID: 23453838]
[13]
Meek EC, Chambers HW, Coban A, et al. Synthesis and in vitro and in vivo inhibition potencies of highly relevant nerve agent surrogates. Toxicol Sci 2012; 126(2): 525-33.
[http://dx.doi.org/10.1093/toxsci/kfs013] [PMID: 22247004]
[14]
Pond AL, Chambers HW, Chambers JE. Organophosphate detoxication potential of various rat tissues via A-esterase and aliesterase activities. Toxicol Lett 1995; 78(3): 245-52.
[http://dx.doi.org/10.1016/0378-4274(95)03327-H] [PMID: 7542808]
[15]
Ametamey SM, Honer M, Schubiger PA. Molecular imaging with PET. Chem Rev 2008; 108(5): 1501-16.
[http://dx.doi.org/10.1021/cr0782426] [PMID: 18426240]
[16]
Neumann KD, Thompson CM, Blecha JE, Gerdes JM, VanBrocklin HF. An improved radiosynthesis of O-(2-[18 F]fluoroethyl)-O-(p-nitrophenyl)methyl-phosphonate: A first-in-class cholinesterase PET tracer. J Labelled Comp Radiopharm 2017; 60(7): 337-42.
[http://dx.doi.org/10.1002/jlcr.3511] [PMID: 28406525]
[17]
Thompson CM, Gerdes JM, VanBrocklin HF. Positron emission tomography studies of organophosphate chemical threats and oxime countermeasures. Neurobiol Dis 2020.133104455
[http://dx.doi.org/10.1016/j.nbd.2019.04.011] [PMID: 31022458]
[18]
Rempel BP, Price EW, Phenix CP. Molecular imaging of hydrolytic enzymes using PET and SPECT. Mol Imaging 2017.161536012117717852
[http://dx.doi.org/10.1177/1536012117717852] [PMID: 28927325]
[19]
Worek F, Aurbek N, Wetherell J, Pearce P, Mann T, Thiermann H. Inhibition, reactivation and aging kinetics of highly toxic organophosphorus compounds: pig versus minipig acetylcholinesterase. Toxicology 2008; 244(1): 35-41.
[http://dx.doi.org/10.1016/j.tox.2007.10.021] [PMID: 18054823]
[20]
Mercey G, Verdelet T, Renou J, et al. Reactivators of acetylcholinesterase inhibited by organophosphorus nerve agents. Acc Chem Res 2012; 45(5): 756-66.
[http://dx.doi.org/10.1021/ar2002864] [PMID: 22360473]
[21]
Jokanović M. Biotransformation of organophosphorus compounds. Toxicology 2001; 166(3): 139-60.
[http://dx.doi.org/10.1016/S0300-483X(01)00463-2] [PMID: 11543910]
[22]
Fukuto TR, Metcalf RL. The effect of structure on the reactivity of alkylphosphonate esters. J Am Chem Soc 1959; 81: 372-7.
[http://dx.doi.org/10.1021/ja01511a025]
[23]
Scott PJH, Hockley BG, Kung HF, Manchanda R, Zhang W, Kilbourn MR. Studies into radiolytic decomposition of fluorine-18 labeled radiopharmaceuticals for positron emission tomography. Appl Radiat Isot 2009; 67(1): 88-94.
[http://dx.doi.org/10.1016/j.apradiso.2008.08.015] [PMID: 18951032]
[24]
Libert LC, Franci X, Plenevaux AR, et al. Production at the Curie level of no-carrier-added 6-18F-fluoro-L-dopa. J Nucl Med 2013; 54(7): 1154-61.
[http://dx.doi.org/10.2967/jnumed.112.112284] [PMID: 23658219]
[25]
Klok RP, Klein PJ, van Erckel BNM, et al. Synthesis of 2-(1,1-dicyanopropen-2-yl)-6-(2[18F]-fluoroethyl)-methylamino-naphthalene ([18F]FDDNP). Appl Radiat Isot 2008; 66: 203-7.
[http://dx.doi.org/10.1016/j.apradiso.2007.10.008] [PMID: 18053733]
[26]
Liu S, Ellars CE, Edwards DS. Ascorbic acid: useful as a buffer agent and radiolytic stabilizer for metalloradiopharmaceuticals. Bioconjug Chem 2003; 14(5): 1052-6.
[http://dx.doi.org/10.1021/bc034109i] [PMID: 13129412]
[27]
Chaubal MV, Kipp J, Rabinow B. Excipient selection and criteria for injectable dosage formsExcipient development for pharmaceutical, biotechnology and drug delivery systems. New York, NY: CRC Press 2006; pp. 271-90.
[http://dx.doi.org/10.1201/9781420004137.ch16]
[28]
Tawfik DS, Eshhar Z, Bentolila A, et al. 1,8-Diazabicyclo[5.4.0]undecene mediated transesterification of p-nitrophenyl phosphonates: a novel route to phosphono esters. Synthesis 1993; 968-72.
[http://dx.doi.org/10.1055/s-1993-25982]
[29]
Loening AM, Gambhir SS. AMIDE: A free software tool for multimodality medical image analysis. Mol Imaging 2003; 2(3): 131-7.
[http://dx.doi.org/10.1162/153535003322556877] [PMID: 14649056]
[30]
Paxinos G. The Rat Nervous System. 4th ed. San Diego, CA: Academic Press 2005.
[31]
Paxinos G, Watson C. The rat brain in stereotaxic coordinates. 6th ed. Boston, MA: Academic Press/Elsevier 2007.
[32]
Walker WFH, Dominique G. Anatomy and Dissection of the Rat. 3rd ed. New York, NY: Freeman, W. H. & Company 1997.
[33]
Gambhir SS. Quantitative assay development for PETMolecular Imaging and its Biological Applications. New York, NY: Springer 2004; pp. 130-2.
[34]
Maxwell DM. The specificity of carboxylesterase protection against the toxicity of organophosphorus compounds. Toxicol Appl Pharmacol 1992; 114(2): 306-12.
[http://dx.doi.org/10.1016/0041-008X(92)90082-4] [PMID: 1609424]
[35]
Maxwell DM, Brecht KM. Carboxylesterase: specificity and spontaneous reactivation of an endogenous scavenger for organophosphorus compounds. J Appl Toxicol 2001; 21(Suppl. 1): S103-7.
[http://dx.doi.org/10.1002/jat.833] [PMID: 11920929]
[36]
Dettbarn WD, Yang ZP, Milatovic D. Different role of carboxylesterases in toxicity and tolerance to paraoxon and DFP. Chem Biol Interact 1999; 119-120: 445-54.
[http://dx.doi.org/10.1016/S0009-2797(99)00057-5] [PMID: 10421482]
[37]
Bahar FG, Ohura K, Ogihara T, Imai T. Species difference of esterase expression and hydrolase activity in plasma. J Pharm Sci 2012; 101(10): 3979-88.
[http://dx.doi.org/10.1002/jps.23258] [PMID: 22833171]
[38]
Song X, Pope C, Murthy R, Shaikh J, Lal B, Bressler JP. Interactive effects of paraoxon and pyridostigmine on blood-brain barrier integrity and cholinergic toxicity. Toxicol Sci 2004; 78(2): 241-7.
[http://dx.doi.org/10.1093/toxsci/kfh076] [PMID: 14976354]
[39]
Gearhart JM, Jepson GW, Clewell HJ, Andersen ME, Conolly RB. Physiologically based pharmacokinetic model for the inhibition of acetylcholinesterase by organophosphate esters. Environ Health Perspect 1994; 102(Suppl. 11): 51-60.
[http://dx.doi.org/10.1289/ehp.94102s1151] [PMID: 7737042]
[40]
Little PJ, Reynolds ML, Bowman ER, Martin BR. Tissue disposition of [3H]sarin and its metabolites in mice. Toxicol Appl Pharmacol 1986; 83(3): 412-9.
[http://dx.doi.org/10.1016/0041-008X(86)90223-1] [PMID: 3705071]
[41]
Martin BR. Biodisposition of [3H]diisopropylfluorophosphate in mice. Toxicol Appl Pharmacol 1985; 77(2): 275-84.
[http://dx.doi.org/10.1016/0041-008X(85)90327-8] [PMID: 3975900]
[42]
Reynolds ML, Little PJ, Thomas BF, Bagley RB, Martin BR. Relationship between the biodisposition of [3H]soman and its pharmacological effects in mice. Toxicol Appl Pharmacol 1985; 80(3): 409-20.
[http://dx.doi.org/10.1016/0041-008X(85)90385-0] [PMID: 4035696]
[43]
Ding SJ, Carr J, Carlson JE, et al. Five tyrosines and two serines in human albumin are labeled by the organophosphorus agent FP-biotin. Chem Res Toxicol 2008; 21(9): 1787-94.
[http://dx.doi.org/10.1021/tx800144z] [PMID: 18707141]
[44]
Peeples ES, Schopfer LM, Duysen EG, et al. Albumin, a new biomarker of organophosphorus toxicant exposure, identified by mass spectrometry. Toxicol Sci 2005; 83(2): 303-12.
[http://dx.doi.org/10.1093/toxsci/kfi023] [PMID: 15525694]
[45]
Kikuchi T, Okamura T, Zhang MR, Fukushi K, Irie T. In vivo evaluation of N-[18F]fluoroethylpiperidin-4ylmethyl acetate in rats compared with MP4A as a probe for measuring cerebral acetylcholinesterase activity. Synapse 2010; 64(3): 209-15.
[http://dx.doi.org/10.1002/syn.20720] [PMID: 19862687]
[46]
Funaki Y, Kato M, Iwata R, et al. Evaluation of the binding characteristics of [5-(11)C-methoxy] Donepezil in the rat brain for in vivo visualization of acetylcholinesterase. J Pharmacol Sci 2003; 91(2): 105-12.
[http://dx.doi.org/10.1254/jphs.91.105] [PMID: 12686754]
[47]
Planas AM, Crouzel C, Hinnen F, et al. Rat brain acetylcholinesterase visualized with [11C]physostig-mine. Neuroimage 1994; 1(3): 173-80.
[http://dx.doi.org/10.1006/nimg.1994.1002] [PMID: 9343568]
[48]
Strauss V, Rey Moreno MC, Vogt J, et al. Acetylcholinesterase measurement in various brain regions and muscles of juvenile, adolescent, and adult rats. Toxicol Mech Methods 2017; 27(9): 666-76.
[http://dx.doi.org/10.1080/15376516.2017.1349849] [PMID: 28671028]
[49]
Phillis JW. Acetylcholine release from the central nervous system: a 50-year retrospective. Crit Rev Neurobiol 2005; 17(3-4): 161-217.
[http://dx.doi.org/10.1615/CritRevNeurobiol.v17.i3-4.30] [PMID: 17341198]
[50]
Segal M, Greenberger V, Israeli M, Biegon A. A correlation between regional acetylcholinesterase activity in rat brain and performance in a spatial task. Behav Brain Res 1988; 30(2): 215-9.
[http://dx.doi.org/10.1016/0166-4328(88)90150-7] [PMID: 3166718]
[51]
Biegon A, Wolff M. Quantitative histochemistry of acetylcholinesterase in rat and human brain postmortem. J Neurosci Methods 1986; 16(1): 39-45.
[http://dx.doi.org/10.1016/0165-0270(86)90006-3] [PMID: 2422501]
[52]
Allen LV Jr, Ansel HC. SolutionsAnsel’s Pharmaceutical Dosage Forms and Drug Delivery Systems. 10th ed. Baltimore, MD: Wolter Kluer Health 2014; pp. 396-444.
[53]
Bittner B, Mountfield RJ. Intravenous administration of poorly soluble new drug entities in early drug discovery: the potential impact of formulation on pharmacokinetic parameters. Curr Opin Drug Discov Devel 2002; 5(1): 59-71.
[PMID: 11865674]
[54]
Shi Y, Porter W, Merdan T, Li LC. Recent advances in intravenous delivery of poorly water-soluble compounds. Expert Opin Drug Deliv 2009; 6(12): 1261-82.
[http://dx.doi.org/10.1517/17425240903307423] [PMID: 19941409]
[55]
Siti HN, Kamisah Y, Kamsiah J. The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (a review). Vascul Pharmacol 2015; 71: 40-56.
[http://dx.doi.org/10.1016/j.vph.2015.03.005] [PMID: 25869516]
[56]
Ashor AW, Lara J, Mathers JC, Siervo M. Effect of vitamin C on endothelial function in health and disease: a systematic review and meta-analysis of randomised controlled trials. Atherosclerosis 2014; 235(1): 9-20.
[http://dx.doi.org/10.1016/j.atherosclerosis.2014.04.004] [PMID: 24792921]
[57]
Marx G. Multiple modes of blood coagulation. J Bone Marrow Res 2014; p. 21000141.
[http://dx.doi.org/10.4172/2329-8820.1000141]
[58]
Tyml K. Critical role for oxidative stress, platelets, and coagulation in capillary blood flow impairment in sepsis. Microcirculation 2011; 18(2): 152-62.
[http://dx.doi.org/10.1111/j.1549-8719.2010.00080.x] [PMID: 21199094]
[59]
Savini I, Catani MV, Arnone R, et al. Translational control of the ascorbic acid transporter SVCT2 in human platelets. Free Radic Biol Med 2007; 42(5): 608-16.
[http://dx.doi.org/10.1016/j.freeradbiomed.2006.11.028] [PMID: 17291984]
[60]
De Neef JH, Jordaan KM, Porsius AJ. The effects of paraoxon on blood pressure in the anaesthetized and in the conscious rat. Br J Pharmacol 1982; 77(2): 223-32.
[http://dx.doi.org/10.1111/j.1476-5381.1982.tb09289.x] [PMID: 7139183]
[61]
Golderman V, Shavit-Stein E, Tamarin I, et al. The organophosphate paraoxon and its antidote obidoxime inhibit thrombin activity and affect coagulation in vitro. PLoS One 2016; 11(9): e0163787
[http://dx.doi.org/10.1371/journal.pone.0163787] [PMID: 27689805]
[62]
Quistad GB, Casida JE. Sensitivity of blood-clotting factors and digestive enzymes to inhibition by organophosphorus pesticides. J Biochem Mol Toxicol 2000; 14(1): 51-6.
[http://dx.doi.org/10.1002/(SICI)1099-0461(2000)14:1<51::AID-JBT7>3.0.CO;2-W] [PMID: 10561082]

© 2024 Bentham Science Publishers | Privacy Policy