7 research outputs found

    Direct Plasma Metabolite Analysis of Positron Emission Tomography Radioligands by Micellar Liquid Chromatography with Radiometric Detection

    No full text
    Determination of radio-metabolites in plasma samples taken during a positron emission tomography (PET) study is an important component in the pharmacokinetic evaluation of PET radioligands. We have developed and validated a new analytical procedure for the plasma metabolite analysis of PET radioligands based on micellar liquid chromatography using an anionic surfactant mobile phase. Chromatographic separation was performed on an octadecyl semipreparative column (10 mm I.D. × 160 mm, 10 μm) using 100 mM sodium dodecyl sulfate (SDS) and 1-butanol in 10 mM sodium-phosphate (pH 7.2) at a flow rate of 5 mL/min. The samples taken from monkey or human plasma during PET measurements were directly injected into a liquid chromatographic (LC) system coupled to an online radiometric detector under micellar conditions using 1–2% (v/v) 1-butanol mobile phase to remove plasma proteins and concentrate the analytes at the column head. At 2 min, mobile phase was changed to elute and separate PET radioligand and its radiometabolites with high peak capacity under high submicellar conditions (10–25% 1-butanol). This procedure allowed direct plasma injection (up to 2 mL) into the LC column without any pretreatment with a short analysis-time of 8–10 min. Satisfactory reproducibility, linearity, sensitivity, accuracy and recovery were obtained in the validation study. The developed method was successfully applied to study the metabolism for diverse groups of PET radioligands and provided reliable determination of PET radioligands in human and monkey plasma. This method is advantageous in terms of simplifying and shortening the processes required to analyze short-lived radioligands as well as in providing a more accurate estimation of the metabolite corrected input function, especially for the radioligands with lower recoveries or degradation potential during the deproteination process in a conventional procedure

    Direct Plasma Metabolite Analysis of Positron Emission Tomography Radioligands by Micellar Liquid Chromatography with Radiometric Detection

    No full text
    Determination of radio-metabolites in plasma samples taken during a positron emission tomography (PET) study is an important component in the pharmacokinetic evaluation of PET radioligands. We have developed and validated a new analytical procedure for the plasma metabolite analysis of PET radioligands based on micellar liquid chromatography using an anionic surfactant mobile phase. Chromatographic separation was performed on an octadecyl semipreparative column (10 mm I.D. × 160 mm, 10 μm) using 100 mM sodium dodecyl sulfate (SDS) and 1-butanol in 10 mM sodium-phosphate (pH 7.2) at a flow rate of 5 mL/min. The samples taken from monkey or human plasma during PET measurements were directly injected into a liquid chromatographic (LC) system coupled to an online radiometric detector under micellar conditions using 1–2% (v/v) 1-butanol mobile phase to remove plasma proteins and concentrate the analytes at the column head. At 2 min, mobile phase was changed to elute and separate PET radioligand and its radiometabolites with high peak capacity under high submicellar conditions (10–25% 1-butanol). This procedure allowed direct plasma injection (up to 2 mL) into the LC column without any pretreatment with a short analysis-time of 8–10 min. Satisfactory reproducibility, linearity, sensitivity, accuracy and recovery were obtained in the validation study. The developed method was successfully applied to study the metabolism for diverse groups of PET radioligands and provided reliable determination of PET radioligands in human and monkey plasma. This method is advantageous in terms of simplifying and shortening the processes required to analyze short-lived radioligands as well as in providing a more accurate estimation of the metabolite corrected input function, especially for the radioligands with lower recoveries or degradation potential during the deproteination process in a conventional procedure

    Direct Plasma Metabolite Analysis of Positron Emission Tomography Radioligands by Micellar Liquid Chromatography with Radiometric Detection

    No full text
    Determination of radio-metabolites in plasma samples taken during a positron emission tomography (PET) study is an important component in the pharmacokinetic evaluation of PET radioligands. We have developed and validated a new analytical procedure for the plasma metabolite analysis of PET radioligands based on micellar liquid chromatography using an anionic surfactant mobile phase. Chromatographic separation was performed on an octadecyl semipreparative column (10 mm I.D. × 160 mm, 10 μm) using 100 mM sodium dodecyl sulfate (SDS) and 1-butanol in 10 mM sodium-phosphate (pH 7.2) at a flow rate of 5 mL/min. The samples taken from monkey or human plasma during PET measurements were directly injected into a liquid chromatographic (LC) system coupled to an online radiometric detector under micellar conditions using 1–2% (v/v) 1-butanol mobile phase to remove plasma proteins and concentrate the analytes at the column head. At 2 min, mobile phase was changed to elute and separate PET radioligand and its radiometabolites with high peak capacity under high submicellar conditions (10–25% 1-butanol). This procedure allowed direct plasma injection (up to 2 mL) into the LC column without any pretreatment with a short analysis-time of 8–10 min. Satisfactory reproducibility, linearity, sensitivity, accuracy and recovery were obtained in the validation study. The developed method was successfully applied to study the metabolism for diverse groups of PET radioligands and provided reliable determination of PET radioligands in human and monkey plasma. This method is advantageous in terms of simplifying and shortening the processes required to analyze short-lived radioligands as well as in providing a more accurate estimation of the metabolite corrected input function, especially for the radioligands with lower recoveries or degradation potential during the deproteination process in a conventional procedure

    Development of [<i>Carbonyl</i>-<sup>11</sup>C]AZ13198083, a Novel Histamine Type‑3 Receptor Radioligand with Favorable Kinetics

    No full text
    The histamine subtype-3 receptor (H<sub>3</sub>R) is implicated in a range of central nervous system disorders, and several radioligands have been developed for H<sub>3</sub>R positron emission tomography imaging. However, a limitation of currently used PET radioligands for H<sub>3</sub>R is the slow binding kinetics in high density brain regions. To address this, we herein report the development of three novel candidate H<sub>3</sub>R radioligands, namely, [<i>carbonyl</i>-<sup>11</sup>C]­AZ13153556 ([<i>carbonyl</i>-<sup>11</sup>C]<b>4</b>), [<i>carbonyl</i>-<sup>11</sup>C]­AZD5213­([<i>carbonyl</i>-<sup>11</sup>C]<b>5</b>), and [<i>carbonyl</i>-<sup>11</sup>C]­AZ13198083 ([<i>carbonyl</i>-<sup>11</sup>C]<b>6</b>), and their subsequent preclinical evaluation in nonhuman primates (NHP). Radioligands [<i>carbonyl</i>-<sup>11</sup>C]<b>4</b>–<b>6</b> were produced and isolated in high radioactivity (>1000 MBq), radiochemical purity (>99%), and moderate molar activity (19–28 GBq/μmol at time of injection) using a palladium-mediated <sup>11</sup>C-aminocarbonylation protocol. All three radioligands showed high brain permeability as well as a regional brain radioactivity distribution in accordance with H<sub>3</sub>R expression (striatum > cortex > cerebellum). [<i>Carbonyl</i>-<sup>11</sup>C]<b>6</b> displayed the most favorable in vivo kinetics and brain uptake, with an early peak in the striatal time–activity curve followed by a progressive washout from the brain. The specificity and on-target kinetics of [<i>carbonyl</i>-<sup>11</sup>C]<b>6</b> were next investigated in pretreatment and displacement studies. After pretreatment or displacement with <b>5</b> (0.1 mg/kg), a uniformly low distribution of radioactivity across the NHP brain was observed. Collectively, this work demonstrates that [<i>carbonyl</i>-<sup>11</sup>C]<b>6</b> is a promising candidate for H<sub>3</sub>R imaging in human subjects

    Development of [<i>Carbonyl</i>-<sup>11</sup>C]AZ13198083, a Novel Histamine Type‑3 Receptor Radioligand with Favorable Kinetics

    No full text
    The histamine subtype-3 receptor (H<sub>3</sub>R) is implicated in a range of central nervous system disorders, and several radioligands have been developed for H<sub>3</sub>R positron emission tomography imaging. However, a limitation of currently used PET radioligands for H<sub>3</sub>R is the slow binding kinetics in high density brain regions. To address this, we herein report the development of three novel candidate H<sub>3</sub>R radioligands, namely, [<i>carbonyl</i>-<sup>11</sup>C]­AZ13153556 ([<i>carbonyl</i>-<sup>11</sup>C]<b>4</b>), [<i>carbonyl</i>-<sup>11</sup>C]­AZD5213­([<i>carbonyl</i>-<sup>11</sup>C]<b>5</b>), and [<i>carbonyl</i>-<sup>11</sup>C]­AZ13198083 ([<i>carbonyl</i>-<sup>11</sup>C]<b>6</b>), and their subsequent preclinical evaluation in nonhuman primates (NHP). Radioligands [<i>carbonyl</i>-<sup>11</sup>C]<b>4</b>–<b>6</b> were produced and isolated in high radioactivity (>1000 MBq), radiochemical purity (>99%), and moderate molar activity (19–28 GBq/μmol at time of injection) using a palladium-mediated <sup>11</sup>C-aminocarbonylation protocol. All three radioligands showed high brain permeability as well as a regional brain radioactivity distribution in accordance with H<sub>3</sub>R expression (striatum > cortex > cerebellum). [<i>Carbonyl</i>-<sup>11</sup>C]<b>6</b> displayed the most favorable in vivo kinetics and brain uptake, with an early peak in the striatal time–activity curve followed by a progressive washout from the brain. The specificity and on-target kinetics of [<i>carbonyl</i>-<sup>11</sup>C]<b>6</b> were next investigated in pretreatment and displacement studies. After pretreatment or displacement with <b>5</b> (0.1 mg/kg), a uniformly low distribution of radioactivity across the NHP brain was observed. Collectively, this work demonstrates that [<i>carbonyl</i>-<sup>11</sup>C]<b>6</b> is a promising candidate for H<sub>3</sub>R imaging in human subjects

    Synthesis of Three Novel Fluorine-18 Labeled Analogues of l-Deprenyl for Positron Emission Tomography (PET) studies of Monoamine Oxidase B (MAO-B)

    No full text
    The aim in this project was to synthesize and to study fluorine-18 labeled analogues of l-deprenyl which bind selectively to the enzyme monoamine oxidase B (MAO-B). Three fluorinated l-deprenyl analogues have been generated in multistep organic syntheses. The most promising fluorine-18 compound <i>N</i>-[(2<i>S</i>)-1-[<sup>18</sup>F]fluoro-3-phenylpropan-2-yl]-<i>N</i>-methylprop-2-yn-1-amine (<b>4c</b>) was synthesized by a one-step fluorine-18 nucleophilic substitution reaction. Autoradiography on human brain tissue sections demonstrated specific binding for compound <b>4c</b> to brain regions known to have a high content of MAO-B. In addition, the corresponding nonradioactive fluorine-19 compound (<b>13</b>) inhibited recombinant human MAO-B with an IC<sub>50</sub> of 170.5 ± 29 nM but did not inhibit recombinant human MAO-A (IC<sub>50</sub> > 2000 nM), demonstrating its specificity. Biodistribution of <b>4c</b> in mice showed high initial brain uptake leveling at 5.2 ± 0.04%ID/g after 2 min post injection. In conclusion, compound <b>4c</b> is a specific inhibitor of MAO-B with high initial brain uptake in mice and is, therefore, a candidate for further investigation in PET

    Discovery and Preclinical Validation of [<sup>11</sup>C]AZ13153556, a Novel Probe for the Histamine Type 3 Receptor

    No full text
    The histamine type 3 receptor (H<sub>3</sub>) is a G protein-coupled receptor implicated in several disorders of the central nervous system. Herein, we describe the radiolabeling and preclinical evaluation of a candidate radioligand for the H<sub>3</sub> receptor, 4-(1<i>S</i>,2<i>S</i>)-2-(4-cyclobutylpiperazine-1-carbonyl)­cyclopropyl]-<i>N</i>-methyl-benzamide (<b>5</b>), and its comparison with one of the frontrunner radioligands for H<sub>3</sub> imaging, namely, GSK189254 (<b>1</b>). Compounds <b>1</b> and <b>5</b> were radiolabeled with tritium and carbon-11 for in vitro and in vivo imaging experiments. The in vitro binding of [<sup>3</sup>H]<b>1</b> and [<sup>3</sup>H]<b>5</b> was examined by (i) saturation binding to rat and nonhuman primate brain tissue homogenate and (ii) in vitro autoradiography on tissue sections from rat, guinea pig, and human brain. The in vivo binding of [<sup>11</sup>C]<b>1</b> and [<sup>11</sup>C]<b>5</b> was examined by PET imaging in mice and nonhuman primates. <i>B</i><sub>max</sub> values obtained from Scatchard analysis of [<sup>3</sup>H]<b>1</b> and [<sup>3</sup>H]<b>5</b> binding were in good agreement. Autoradiography with [<sup>3</sup>H]<b>5</b> on rat, guinea pig, and human brain slices showed specific binding in regions known to be enhanced in H<sub>3</sub> receptors, a high degree of colocalization with [<sup>3</sup>H]<b>1</b>, and virtually negligible nonspecific binding in tissue. PET measurements in mice and nonhuman primates demonstrated that [<sup>11</sup>C]<b>5</b> binds specifically and reversibly to H<sub>3</sub> receptors in vivo with low nonspecific binding in brain tissue. Whereas [<sup>11</sup>C]<b>1</b> showed similar binding characteristics in vivo, the binding kinetics appeared faster for [<sup>11</sup>C]<b>5</b> than for [<sup>11</sup>C]<b>1</b>. Conclusions: [<sup>11</sup>C]<b>5</b> has suitable properties for quantification of H<sub>3</sub> receptors in nonhuman primate brain and has the potential to offer improved binding kinetics in man compared to [<sup>11</sup>C]<b>1</b>
    corecore