PET radiotracers: crossing the blood-brain barrier and surviving metabolism

Victor W Pike, Victor W Pike

Abstract

Radiotracers for imaging protein targets in the living human brain with positron emission tomography (PET) are increasingly useful in clinical research and in drug development. Such radiotracers must fulfill many criteria, among which an ability to enter brain adequately and reversibly without contamination by troublesome radiometabolites is desirable for accurate measurement of the density of a target protein (e.g. neuroreceptor, transporter, enzyme or plaque). Candidate radiotracers can fail as a result of poor passive brain entry, rejection from brain by efflux transporters or undesirable metabolism. These issues are reviewed. Emerging PET radiotracers for measuring efflux transporter function and new strategies for ameliorating radiotracer metabolism are discussed. A growing understanding of the molecular features affecting the brain penetration, metabolism and efflux transporter sensitivity of prospective radiotracers should ultimately lead to their more rational and efficient design, and also to their greater efficacy.

Figures

Figure 1
Figure 1
PET Radiotracers: crossing the blood-brain barrier and surviving metabolism. The cartoon represents a voxel in brain, with edge dimensions of a millimeter or so, as seen with a PET camera after intravenous administration of a radiotracer. Blood represents about 5% of the total volume. The red circles indicate some of the radioactive entities that might be generated from the parent radiotracer (Rf) and their locations. The green areas represent efflux transporters at the blood-brain barrier (e.g., P-gp). Key: R = radiotracer; M = radiometabolite. Subscripts: f = free; b = plasma protein-bound; np = non BBB-penetrant; ns = non-specifically bound to brain; sp = specifically bound to brain. Radiotracer development for a protein target within brain aims to achieve ready passive diffusion of the radiotracer across the BBB, without impediment by efflux transporters, to give a high ratio of specifically bound radiotracer (Rsp) to non-specifically bound (Rnp) plus free (Rf) radiotracer, while suppressing the presence of radiometabolite entities in brain.
Figure 2
Figure 2
[11C]WAY-110635 for imaging brain 5-HT1A receptors: avoiding troublesome radiometabolites by judicious choice of position of radiolabel. WAY-100635 is metabolized in the periphery by hydrolysis of its amide bond. When WAY-100635 is labeled with carbon-11 in its O-methyl group, metabolism produces [11C]A as a major radiometabolite in plasma. [11C]A is able to enter brain to bind non-specifically, and also potentially specifically to some proteins for which it has moderately high affinity (5-HT1A receptors and α1 adrenoceptors) [54]. This radiometabolite therefore attenuates the signal from the unchanged radiotracer, [O-methyl-11C]WAY-100635, and exacerbates biomathematical analysis. By placing the radiolabel in the carbonyl position, metabolism of the radiotracer, [carbonyl-11C]WAY-100635, to [11C]A is avoided. The major radiometabolite becomes [11C]cyclohexanecarboxylate anion, which has only low transient entry into brain [55]. Consequently, with this radiotracer a much greater proportion of the radioactivity in brain is specifically bound unchanged radioligand [27], and biomathematical modeling of the acquired PET data is possible to deliver measures of receptor density [79, 80].

References

    1. Jacobs AH, et al. PET-based molecular imaging in neuroscience. Eur J Nucl Med & Mol Imaging. 2003;30:1051–1065.
    1. Lee CM, Farde L. Using positron emission tomography to facilitate drug development. TiPs. 2006;27:310–316.
    1. Fowler JS, Ido T. In: Handbook of Radiopharmaceuticals, Radiochemistry and Applications. Welch MJ, Redvanly CS, editors. Chapter 9. Wiley; Chichester: 2003. pp. 307–321.
    1. Pike VW. Positron-emitting radioligands for studies in vivo Probes for human psychopharmacology. J Psychopharmacology. 1993;7:139–158.
    1. Laruelle M, et al. Relationships between radiotracer properties and image quality in molecular imaging of the brain with positron emission tomography. Mol Imaging Biol. 2003;5:363–375.
    1. Lee G, Bendayan R. Functional expression and localization of P-glycoprotein in the central nervous system: relevance to the pathogenesis and treatment of neurological disorders. Pharm Res. 2004;21:1313–1330.
    1. Dallas S, et al. Multidrug resistance-associated proteins: expression and function in the central nervous system. Pharmacol Rev. 2006;58:140–161.
    1. Innis RB, et al. Concensus nomenclature for in vivo imaging of reversibly binding radioligands. J Cerebr Blood Flow Metab. 2007;27:1533–1539.
    1. Raub TJ. P-glycoprotein recognition of substrates and circumvention through rational drug design. Mol Pharmaceutics. 2006;3:3–25.
    1. Fischer H, et al. Blood-brain barrier permeation: molecular parameters governing passive diffusion. J Membrane Biol. 1998;165:201–211.
    1. Gerebtzoff G, Seelig A. In silico prediction of blood-brain barrier permeation using the calculated molecular cross-sectional area as main parameter. J Chem Inf Model. 2006;46:2638–2650.
    1. Seelig A. The role of size and charge for blood-brain barrier permeation of drugs and fatty acids. J Mol Neurosci. 2007;33:323–341.
    1. Waterhouse RN. Determination of lipophilicity and its use as a predictor of blood-brain barrier penetration of molecular imaging agents. Mol Imaging Biol. 2003;5:376–389.
    1. Donohue SR, et al. Synthesis, ex vivo evaluation and radiolabeling of potent 1,5-diphenyl-pyrrolidin-2-one cannabinoid subtype-1 (CB1) receptor ligands as candidates for in vivo imaging. J Med Chem. 2008;51:5833–5842.
    1. Yasuno F, et al. The PET radioligand [11C]MePPEP binds reversibly and with high specific signal to cannabinoid CB1 receptors in nonhuman primate brain. Neuropsychopharmacology. 2008;33:259–269.
    1. Terry GE, et al. Evaluation of an inverse agonist with high affinity for the CB1 receptor, [11C]MePPEP, in rodents and humans. NeuroImage. 2008;41 (Suppl 2):T197.
    1. Elsinga PH, et al. PET studies on P-glycoprotein function in the blood-brain barrier: how it affects uptake and binding of drugs within the CNS. Curr Pharm Design. 2004;10:1493–1503.
    1. Ishiwata K, et al. In vivo evaluation of P-glycoprotein modulation of 8 PET radioligands used clinically. J Nucl Med. 2007;48:81–87.
    1. Syvänen S, et al. Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Disposition. 2009;37:635–643.
    1. Elsinga PH, et al. Positron emission tomography studies on binding of central nervous system drugs and P-glycoprotein function in the rodent brain. Mol Imaging Biol. 2005;7:37–44.
    1. Liow JS, et al. Effect of a P-glycoprotein inhibitor, cyclosporin A, on the disposition in rodent brain and blood of the 5-HT1A receptor radioligand, [11C](R)-(−)-RWAY. Synapse. 2007;61:96–105.
    1. Passchier J, et al. Influence of P-glycoprotein on brain uptake of [18F]MPPF in rats. Eur J Pharmacol. 2000;407:273–280.
    1. Lacan G, et al. Cyclosporine, a P-glycoprotein modulator, increases [18F]MPPF uptake in rat brain and peripheral tissues: microPET and ex vivo studies. Eur J Nucl Med & Mol Imaging. 2008;35:2256–2266.
    1. Farde L, et al. PET characterisation of [carbonyl-11C]WAY-100635 binding to 5-HT1A receptors in the primate brain. Psychopharmacology. 1997;133:196–202.
    1. Yasuno F, et al. Quantification of serotonin 5-HT1A receptors in monkey brain with [11C](−)-RWAY. Synapse. 2006;60:510–520.
    1. Shiue CY, et al. p-[18F]-MPPF: a potential radioligand for PET studies of 5-HT1A receptors in humans. Synapse. 1997;25:147–154.
    1. Pike VW, et al. Exquisite delineation of 5-HT1A receptors in human brain with PET and [carbonyl-11C]WAY-100635. Eur J Pharmacol. 1996;301:R5–R7.
    1. Zhang XY, et al. Quantification of serotonin 5-HT1A receptors in humans with [11C](R)-(−)-RWAY: radiometabolite(s) likely confound brain measurements. Synapse. 2007;61:469–477.
    1. Passchier J, et al. In vivo delineation of 5-HT1A receptors in human brain with p-[18F]MPPF. J Nucl Med. 2000;41:1830–1835.
    1. Doze P, et al. Enhanced cerebral uptake of receptor ligands by modulation of P-glycoprotein function in the blood brain barrier. Synapse. 2000;36:66–74.
    1. Lazarova N, et al. Synthesis and evaluation of [N-methyl-11C]N-desmethyl-loperamide as a new and improved PET radiotracer for imaging P-gp function. J Med Chem. 2008;51:6034–6043.
    1. Summerfield SG, et al. Improving the in vitro prediction of in vivo central nervous system penetration: integrating permeability, P-glycoprotein efflux, and free fractions in blood and brain. J Pharmacol Exp Ther. 2006;316:1282–1290.
    1. Summerfield SG, et al. Toward an improved prediction of human in vivo brain penetration. Xenobiotica. 2008;38:1518–1535.
    1. Willemsen ATM, van den Hoff J. Fundamentals of quantitative PET data analysis. Curr Pharm Design. 2002;8:1513–1526.
    1. Ikoma Y, et al. Quantitative analysis of [11C]verapamil transfer at the human blood-brain barrier for evaluation of P-glycoprotein function. J Nucl Med. 2006;47:1531–1537.
    1. Luurtsma G, et al. Evaluation of (R)-[11C]verapamil as PET tracer of P-glycoprotein function at the blood-brain barrier: kinetics and metabolism in the rat. Nucl Med Biol. 2005;32:87–93.
    1. Pauli-Magnus C, et al. Characterization of the major metabolites of verapamil as substrates and inhibitors of P-glycoprotein. J Pharmacol Exp Ther. 2000;293:376–382.
    1. Zoghbi SS, et al. 11C-Loperamide and its N-desmethyl radiometabolite are avid substrates for brain P-glycoprotein efflux. J Nucl Med. 2008;49:649–656.
    1. Liow JS, et al. P-glycoprotein function at the blood-brain barrier imaged in monkey with 11C-N-desmethyl-loperamide. J Nucl Med. 2009;50:108–115.
    1. Seneca N, et al. Human brain imaging and radiation dosimetry of 11C-N-desmethyl-loperamide, a positron emission tomography radioligand to measure the function of P-glycoprotein. J Nucl Med. 2009;50:807–813.
    1. Okamura T, et al. Noninvasive and quantitative assessment of the function of multidrug resistance-associated protein 1 in the living brain. J Cerebr Blood Flow Metab. 2009;29:504–511.
    1. Zoghbi SS, et al. PET imaging of the dopamine transporter with [18F]FECNT: a polar radioactive metabolite confounds brain activity measurements. J Nucl Med. 2006;47:520–527.
    1. Shetty HU, et al. Identification and regional distribution in rat brain of radiometabolites of the dopamine transporter PET radioligand, [11C]PE2I. Eur J Nucl Med & Mol Imaging. 2007;34:667–678.
    1. Hirvonen J, et al. Measurement of striatal and extrastriatal dopamine transporter binding with high-resolution PET and [11C]PE2I: quantitative modeling and test-retest reproducibility. J Cerebr Blood Flow Metab. 2008;28:1059–1069.
    1. McCarron JA, et al. Synthesis and preliminary evaluation of [11C](R)-RWAY in monkey a new simply labeled PET radioligand for imaging brain 5-HT1A receptors. Eur J Nucl Med & Mol Imaging. 2007;34:1670–1682.
    1. Briard E, et al. Single-step high-yield radiosynthesis and evaluation of a sensitive 18F-labeled ligand for imaging brain peripheral benzodiazepine receptors with PET. J Med Chem. 2009;52:688–699.
    1. Fujimura Y, et al. Quantification of peripheral benzodiazepine receptors in human brain with positron emission tomography and 18F-PBR06. J Nucl Med (In press)
    1. Siméon F, et al. Synthesis and simple 18F-labeling of a high affinity 2-(fluoromethyl)thiazole derivative ([18F]SP203) as a radioligand for imaging brain metabotropic glutamate subtype-5 receptors with PET. J Med Chem. 2007;50:3256–3266.
    1. Brown AK, et al. Metabotropic glutamate subtype-5 (mGluR5) receptors quantified in human brain with a novel radioligand for positron emission tomography. J Nucl Med. 2008;49:2042–2048.
    1. Shetty HU, et al. 3-Fluoro-5-(2-(2-([18F]fluoromethyl)thiazol-4-yl)ethynyl)benzonitrile ([18F]SP203), a radioligand for imaging brain mGluR5 in vivo, is defluorinated by glutathionylation in rat brain. J Pharmacol Exp Therapeutics. 2008;327:727–735.
    1. Gatley SJ, et al. Studies with differentially labeled [11C]cocaine, [11C]norcocaine, [11C]benzoylecgonine, and [11C] and 4′-[18F]fluorococaine to probe the extent to which [11C]cocaine metabolites contribute to PET images of the baboon brain. J Neurochem. 1994;62:1154–1162.
    1. Odano I, et al. [18F]Flumazenil binding to central benzodiazepine receptor studies by PET Quantitative analysis and comparisons with [11C]flumazenil. NeuroImage. 2009;45:891–902.
    1. Pike VW, et al. Pre-clinical development of a radioligand for studies of central 5-HT1A receptors in vivo [11C]WAY-100635. Med Chem Res. 1995;5:208–227.
    1. Osman S, et al. Characterization of the radioactive metabolites of the 5-HT1A receptor radioligand, [O-methyl-11C]WAY-100635, in monkey and human plasma by HPLC Comparison of the behaviour of an identified radioactive metabolite with parent radioligand in monkey using PET. Nucl Med Biol. 1996;23:627–634.
    1. Osman S, et al. Characterisation of the appearance of radioactive metabolites in monkey and human plasma from the 5-HT1A receptor radioligand, [carbonyl-11C]WAY-100635 Explanation of high signal in PET and an aid to biomathematical modelling. Nucl Med Biol. 1998;25:215–223.
    1. Hashimoto K, et al. Deuterium-isotope effect of [11C]N,N-dimethylphenethylamine-α,α-d2 reduction in metabolic trapping rate in brain. Nucl Med Biol. 1986;13:79–80.
    1. Fowler JS, et al. Mechanistic positron emission tomography studies: demonstration of a deuterium isotope effect in the monoamine oxidase-catalyzed binding of [11C]L-deprenyl in living baboon brain. J Neurochem. 1988;51:1524–1534.
    1. Schou M, et al. PET evaluation of novel radiofluorinated reboxetine analogs as norepeienphrine transporter probes in the monkey brain. Synapse. 2004;53:57–67.
    1. Wilson AA, et al. Radiosynthesis and initial evaluation of [18F]FEPPA for PET imaging of peripheral benzodiazepine receptors. Nucl Med Biol. 2008;35:305–314.
    1. Briard E, et al. Synthesis and evaluation in monkey of two sensitive 11C-labeled aryloxyanilide ligands for imaging brain peripheral benzodiazepine receptors in vivo. J Med Chem. 2008;51:17–30.
    1. Tipre DN, et al. PET imaging of brain 5-HT1A receptors in rat in vivo with [18F]FCWAY and improvement by successful inhibition of defluorination with miconazole. J Nucl Med. 2006;47:1–9.
    1. Ryu YH, et al. Disulfiram inhibits defluorination of [18F]FCWAY, reduces bone radioactivity, and enhances visualization of radioligand binding to serotonin 5-HT1A receptors in human brain. J Nucl Med. 2007;48:1154–1161.
    1. Matusch A, et al. Metabolism of the A1 adenosine receptor PET ligand [18F]CPFPX by CYP1A2: implications of bolus/infusion studies. Nucl Med Biol. 2006;33:891–898.
    1. Bier D, et al. Metabolism of the A1 adenosine receptor positron emission tomography ligand [18F]8-cyclopentyl-3(-3-fluoropropyl)-1-propylxanthine ([18F]CPFPX) in rodents and humans. Drug Metab Disposition. 2006;34:570–576.
    1. Ding YS, et al. 6-[18F]Fluoro-A-85380, a new PET tracer for the nicotonic acetylcholine receptor: studies in the human brain and in vivo demonstration of specific binding in white matter. Synapse. 2004;53:184–189.
    1. Rinne JO, et al. PET examination of the monoamine transporter with [11C]β-CIT and [11C]β-CFT in early Parkinson’s disease. Synapse. 1995;21:97–103.
    1. Yasuno F, et al. PET Imaging of neurokinin-1 receptors with [18F]SPA-RQ in human subjects: assessment of reference tissue models and their test-test reproducibility. Synapse. 2007;61:242–251.
    1. Mukherjee J, et al. Brain imaging of 18F-fallypride in normal volunteers: blood analysis, distribution, test-retest studies, and preliminary assessment of sensitivity to aging effects on dopamine D-2/D-3 receptors. Synapse. 2002;46:170–188.
    1. Jones AKP, et al. Quantification of [11C]diprenorphine cerebral kinetics in man acquired by PET using presaturation, pulse-chase and tracer only protocols. J Neurosci Methods. 1994;51:123–134.
    1. DaSilva JN, et al. Imaging cAMP-specific phosphodiesterases-4 in human brain with R-[11C]rolipram and positron emission tomography. Eur J Nucl Med. 2002;29:1680–1683.
    1. Farde L, et al. Subsituted benzamides as ligands for visualization of dopamine receptor binding in the human brain by positron emission tomography. Proc Natl Acad Sci USA. 1985;82:3863–3867.
    1. Andrée B, et al. The PET radioligand [carbonyl-11C]desmethyl-WAY-100635 binds to 5-HT1A receptors and provides a higher radioactive signal than [carbonyl-11C]WAY-100635 in the human brain. J Nucl Med. 2002;43:292–303.
    1. Hinz R, et al. Validation of a tracer kinetic model for the quantification of 5-HT2A receptors in human brain with [11C]MDL 100,907. J Cerebr Blood Flow Metab. 2007;27:161–172.
    1. Fujita M, et al. Kinetic analysis in healthy humans of a novel positron emission tomography radioligand to image the peripheral benzodiazepine receptor, a potential marker for inflammation. NeuroImage. 2008;40:43–52.
    1. Klunk WE. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh compound-B. Ann Neurol. 2004;55:306–319.
    1. Ginovart N, et al. Positron emission tomography quantification of [11C]-DASB binding to the human serotonin transporter: modeling strategies. J Cerebr Blood Flow Metab. 2001;21:1342–1353.
    1. Kropholler MA, et al. Development of a tracer kinetic plasma input model for (R)-[11C]PK11195 brain studies. J Cerebr Blood Flow Metab. 2005;25:842–851.
    1. Halldin C, et al. Carbon-11-NNC 112: a radioligand for PET examination of striatal and neocortical D1-dopamine receptors. J Nucl Med. 1998;39:2061–2068.
    1. Gunn RN, et al. Tracer kinetic modeling of the 5-HT1A receptor ligand [carbonyl-11C]WAY-100635. NeuroImage. 1998;8:426–440.
    1. Farde L, et al. Quantitative analyses of [carbonyl-11C]WAY-100635 binding to central 5-hydroxytryptamine 1A receptors in man. J Nucl Med. 1998;39:1965–1971.

Source: PubMed

3
Tilaa