ISOTOPIC CARBON CHOLINE ANALOGS

Abstract
Novel choline-derived radiotracer (s) having an isotopic carbon for Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) imaging of disease states related to altered choline metabolism (e.g., tumor imaging of prostate, breast, brain, esophageal, ovarian, endometrial, lung and prostate cancer—primary tumor, nodal disease or metastases).
Description
FIELD OF THE INVENTION

The present invention describes a novel radiotracer(s) for Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) imaging of disease states related to altered choline metabolism (e.g., tumor imaging of prostate, breast, brain, esophageal, ovarian, endometrial, lung and prostate cancer—primary tumor, nodal disease or metastases). The present invention also describes intermediate(s), precursor(s), pharmaceutical composition(s), methods of making, and methods of use of the novel radiotracer(s).


DESCRIPTION OF RELATED ART

The biosynthetic product of choline kinase (EC 2.7.1.32) activity, phosphocholine, is elevated in several cancers and is a precursor for membrane phosphatidylcholine (Aboagye, E. O., et al., Cancer Res 1999; 59:80-4; Exton, J. H., Biochim Biophys Acta 1994; 1212:26-42; George, T. P., et al., Biochim Biophys Acta 1989; 104:283-91; and Teegarden, D., et al., J Biol Chem 1990; 265(11):6042-7). Over-expression of choline kinase and increased enzyme activity have been reported in prostate, breast, lung, ovarian and colon cancers (Aoyama, C., et al., Prog Lipid Res 2004; 43(3):266-81; Glunde, K., et al., Cancer Res 2004; 64(12):4270-6; Glunde, K., et al., Cancer Res 2005; 65(23): 11034-43; Iorio, E., et al., Cancer Res 2005; 65(20): 9369-76; Ramirez de Molina, A., et al., Biochem Biophys Res Commun 2002; 296(3): 580-3; and Ramirez de Molina, A., et al., Lancet Oncol 2007; 8(10): 889-97) and are largely responsible for the increased phosphocholine levels with malignant transformation and progression; the increased phosphocholine levels in cancer cells are also due to increased breakdown via phospholipase C (Glunde, K., et al., Cancer Res 2004; 64(12):4270-6).


Because of this phenotype, together with reduced urinary excretion, [11C]choline has become a prominent radiotracer for positron emission tomography (PET) and PET-Computed Tomography (PET-CT) imaging of prostate cancer, and to a lesser extent imaging of brain, esophageal, and lung cancer (Hara, T., et al., J Nucl Med 2000; 41:1507-13; Hara, T., et al., J Nucl Med 1998; 39:990-5; Hara, T., et al., J Nucl Med 1997; 38:842-7; Kobori, O., et al., Cancer Cell 1999; 86:1638-48; Pieterman, R. M., et al., J Nucl Med 2002; 43(2):167-72; and Reske, S. N. Eur J Nucl Med Mol Imaging 2008; 35:1741). The specific PET signal is due to transport and phosphorylation of the radiotracer to [11C]phosphocholine by choline kinase.


Of interest, however, is that [11C]choline (as well as the fluoro-analog) is oxidized to [11C]betaine by choline oxidase (see FIG. 1 below) (EC 1.1.3.17) mainly in kidney and liver tissues, with metabolites detectable in plasma soon after injection of the radiotracer (Roivainen, A., et al., European Journal of Nuclear Medicine 2000; 27:25-32). This makes discrimination of the relative contributions of parent radiotracer and catabolites difficult when a late imaging protocol is used.




embedded image


[18F]Fluoromethylcholine ([18F]FCH):



embedded image


was developed to overcome the short physical half-life of carbon-11 (20.4 min) (DeGrado, T. R., et al., Cancer Res 2001; 61(1): 110-7) and a number of PET and PET-CT studies with this relatively new radiotracer have been published (Beheshti, M., et al., Eur J Nucl Med Mol Imaging 2008; 35(10): 1766-74; Cimitan, M., et al., Eur J Nucl Med Mol Imaging 2006; 33(12):1387-98; de Jong, I. J., et al., Eur J Nucl Med Mol Imaging 2002; 29:1283-8; and Price, D. T., et al., J Urol 2002; 168(1):273-80). The longer half-life of fluorine-18 (109.8 min) was deemed potentially advantageous in permitting late imaging of tumors when sufficient clearance of parent tracer in systemic circulation had occurred (DeGrado, T. R., et al., J Nucl Med 2002; 43(1):92-6).


WO2001/82864 describes 18F-labeled choline analogs, including [18F]Fluoromethylcholine ([18F]-FCH) and their use as imaging agents (e.g., PET) for the non-invasive detection and localization of neoplasms and pathophysiologies influencing choline processing in the body (Abstract). WO2001/82864 also describes 18F-labeled di-deuterated choline analogs such as [18F]fluoromethyl-[1-2H2]choline ([18F]FDC) (hereinafter referred to as “[18F]D2-FCH”):




embedded image


The oxidation of choline under various conditions; including the relative oxidative stability of choline and [1,2-2H4]choline has been studied (Fan, F., et al., Biochemistry 2007, 46, 6402-6408; Fan, F., et al., Journal of the American Chemical Society 2005, 127, 2067-2074; Fan, F., et al., Journal of the American Chemical Society 2005, 127, 17954-17961; Gadda, G. Biochimica et Biophysica Acta 2003, 1646, 112-118; Gadda, G., Biochimica et Biophysica Acta 2003, 1650, 4-9). Theoretically the effect of the extra deuterium substitution was found to be neglible in the context of a primary isotope effect of 8-10 since the β-secondary isotope effect is ˜1.05 (Fan, F., et al., Journal of the American Chemical Society 2005, 127, 17954-17961).


[18F]Fluoromethylcholine is now used extensively in the clinic to image tumour status (Beheshti, M., et al., Radiology 2008, 249, 389-90; Beheshti, M., et al., Eur J Nucl Med Mol Imaging 2008, 35, 1766-74).


The present invention, as described below, provides a novel 11C-radiolabeled radiotracer that can be used for PET imaging of choline metabolism and exhibits increased metabolic stability and a favourable urinary excretion profile.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts the chemical structures of major choline metabolites and their pathways.



FIG. 3 shows NMR analysis of tetradeuterated choline precursor. Top, 1H NMR spectrum; bottom, 13C NMR spectrum. Both spectra were acquired in CDCl3.



FIG. 4 depicts the HPLC profiles for the synthesis of [18F]fluoromethyl tosylate (9) and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) showing (A) radio-HPLC profile for synthesis of (9) after 15 mins; (B) UV (254 nm) profile for synthesis of (9) after mins; (C) radio-HPLC profile for synthesis of (9) after 10 mins; (D) radio-HPLC profile for crude (9); (E) radio-HPLC profile of formulated (9) for injection; (F) refractive index profile post formulation (cation detection mode).



FIG. 5
a is a picture of a fully assembled cassette of the present invention for the production of [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) via an unprotected precursor.



FIG. 5
b is a picture of a fully assembled cassette of the present invention for the production of [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) via a PMB-protected precursor.



FIG. 6 depicts representative radio-HPLC analysis of potassium permanganate oxidation study. Top row are control samples for [18F]fluoromethylcholine ([18F]FCH) and [18F]fluoromethyl-[1,2-2H4]choline ([18F]D4-FCH), extracts from the reaction mixture at time zero (0 min). Bottom row are extracts after treatment for 20 mins. Left hand side are for [18F]fluoromethylcholine ([18F]FCH), right are for [18F]fluoromethyl-[1,2-2H4]choline ([18F]D4-FCH).



FIG. 7 shows chemical oxidation potential of [18F]fluoromethylcholine and [18F]fluoromethyl-[1,2-2H4]choline in the presence of potassium permanganate.



FIG. 8 shows time-course stability assay of [18F]fluoromethylcholine and [18F]fluoromethyl-[1,2-2H4]choline in the presence of choline oxidase demonstrating conversion of parent compounds to their respective betaine analogues.



FIG. 9 shows representative radio-HPLC analysis of choline oxidase study. Top row are control samples for [18F]fluoromethylcholine and [18F]fluoromethyl-[1,2-2H4]choline, extracts from the reaction mixture at time zero (0 min). Bottom row are extracts after treatment for 40 mins. Left hand side are of [18F]fluoromethylcholine, right are of [18F]fluoromethyl-[1,2-2H4]choline.



FIG. 10. Top: Analysis of the metabolism of [18F]fluoromethylcholine (FCH) to [18F]FCH-betaine and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) to [18F]D4-FCH-betaine by radio-HPLC in mouse plasma samples obtained 15 min after injecting the tracers i.v. into mice. Bottom: summary of the conversion of parent tracers, [18F]fluoromethylcholine (FCH) and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH), to metabolites, [18F]FCH-betaine (FCHB) and [18F]D4-FCH betaine (D4-FCHB), in plasma.



FIG. 11. Biodistribution time course of [18F]fluoromethylcholine (FCH), [18F]fluoromethyl-[1-2H2]choline (D2-FCH) and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) in HCT-116 tumor bearing mice. Inset: the time points selected for evaluation. A) Biodistribution of [18F]fluoromethylcholine; B) biodistribution of [18F]fluoromethyl-[1-2H2]choline; C) biodistribution of [18F]fluoromethyl-[1,2-2H4]choline; D) time course of tumor uptake for [18F]fluoromethylcholine (FCH), [18F]fluoromethyl-[1-2H2]choline (D2-FCH) and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) from charts A-C. Approximately 3.7 MBq of [18F]fluoromethylcholine (FCH), [18F]fluoromethyl-[1-2H2]choline (D2-FCH) and [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) injected into awake male C3H-Hej mice which were sacrificed under isofluorane anesthesia at the indicated time points.



FIG. 12 shows radio-HPLC chromatograms to show distribution of choline radiotracer metabolites in tissue harvested from normal white mice at 30 min p.i. Top row, radiotracer standards; middle row, kidney extracts; bottom row, liver extracts. On the left is [18F]FCH, on the right [18F]D4-FCH.



FIG. 13 show radio-HPLC chromatograms to show metabolite distribution of choline radiotracers in HCT116 tumors 30 min post-injection. Top-row, neat radiotracer standards; bottom row, 30 min tumor extracts. Left side, [18F]FCH; middle, [18F]D4-FCH; right, [11C]choline.



FIG. 14 shows radio-HPLC chromatograms for phosphocholine HPLC validation using HCT116 cells. Left, neat [18F]FCH standard; middle, phosphatase enzyme incubation; right, control incubation.



FIG. 15 shows distribution of radiometabolites for [18F]fluoromethylcholine analogs: 18F]fluoromethylcholine, [18F]fluoromethyl-[1-2H2]choline and [18F]fluoromethyl-[1,2-2H4]choline at selected time points.



FIG. 16 shows tissue profile of [18F]FCH and [18F]D4-FCH. (a) Time versus radioactivity curve for the uptake of [18F]FCH in liver, kidney, urine (bladder) and muscle derived from PET data, and (b) corresponding data for [18F]D4-FCH. Results are the mean±SE; n=4 mice. For clarity upper and lower error bars (SE) have been used. (Leyton, et al., Cancer Res 2009: 69:(19), pp 7721-7727).



FIG. 17 shows tumor profile of [F]FCH and [18F]D4-FCH in SKMEL28 tumor xenograft. (a) Typical [18F]FCH-PET and [18F]D4-FCH-PET images of SKMEL28 tumor-bearing mice showing 0.5 mm transverse sections through the tumor and coronal sections through the bladder. For visualization, 30 to 60 min summed image data are displayed. Arrows point to the tumors (T), liver (L) and bladder (B). (b). Comparison of time versus radioactivity curves for [18F]FCH and [18F]D4-FCH in tumors. For each tumor, radioactivity at each of 19 time frames was determined. Data are mean % ID/vox60 mean±SE (n=4 mice per group). (c) Summary of imaging variables. Data are mean±SE, n=4; *P=0.04. For clarity upper and lower error bars (SE) have been used.



FIG. 18 shows the effect of PD0325901, a mitogenic extracellular kinase inhibitor, on uptake of [18F]D4-FCH in HCT116 tumors and cells. (a) Normalized time versus radioactivity curves in HCT116 tumors following daily treatment for 10 days with vehicle or 25 mg/kg PD0325901. Data are the mean±SE; n=3 mice. (b) Summary of imaging variables % ID/vox60, % ID/vox60max, and AUC. Data are mean±SE; * P=0.05. (c) Intrinsic cellular effect of PD0325901 (1 μM) on [18F]D4-FCH phosphocholine metabolism after treating HCT116 cells for 1 hr with [18F]D4-FCH in culture. Data are mean±SE; n=3; * P=0.03.



FIG. 19 shows expression of choline kinase A in HCT116 tumors. (a) A typical Western blot demonstrating the effect of PD0325901 on tumor choline kinase A (CHKA) protein expression. HCT116 tumors from mice that were injected with PD0325901 (25 mg/kg daily for 10 days, orally) or vehicle were analyzed for CHKA expression by western blotting. β-actin was used as the loading control. (b) Summary densitometer measurements for CHKA expression expressed as a ratio to β-actin. The results are the mean ratios±SE; n=3, * P=0.05.



FIG. 20 shows biodistribution time course of 11C-choline, 11C-D4-choline and 18F-D4-choline in BALB/c nude mice. Approximately 18.5 MBq of 11C-labeled tracer or 3.7 MBq of 18F was administered i.v. into anaesthetized animals prior to sacrifice at indicated time points. Tissues were excised, weighed and counted, with counts normalized to injected dose/g wet weight tissue. Mean values (n=3) and SEM are shown.



FIG. 21 shows metabolic profile of 11C-choline, 11C-D4-choline and 18F-D4-choline in the liver (A) and kidney (B) of BALB/c nude mice. Radiolabelled metabolite profile was assessed at 2, 15, 30 and 60 min after i.v. injection of parent radiotracers using radio-HPLC. Mean values (n=3) and SEM are shown. Abbreviations: Bet-ald, betaine aldehyde; p-Choline, phosphocholine.



FIG. 22 shows metabolic profile of 11C-choline, 11C-D4-choline and 18F-D4-choline in HCT116 tumors. Radiolabelled metabolite profile in HCT116 tumor xenografts was assessed at 15 min and 60 min after i.v. injection of parent radiotracers using radio-HPLC. Mean values (n=3) and SEM are shown. * P<0.05; ** P<0.01; *** P<0.001.



FIG. 23 depicts 11C-choline (◯), 11C-D4-choline (▴) and 18F-D4-choline (▪) PET image analysis. HCT116 tumor uptake profiles were examined following 60 min dynamic PET imaging. A, representative axial PET-CT images of HCT116 tumor-bearing mice (30-60 min summed activity) for 11C-choline, 11C-D4-choline and 18F-D4-choline. Tumor margins, indicated from CT image, are outlined in red. B, The tumor time versus radioactivity curve (TAC). Mean±SEM (n=4 mice per group).



FIG. 24 shows pharmacokinetics of 11C-choline, 11C-D4-choline and 18F-D4-choline in HCT116 tumors. A, Modified compartmental modeling analysis, taking into account plasma metabolites and their flux into the exchangeable space in tumor, was used to derive Ki, a measure of irreversible retention within the tumor. B, The kinetic parameter, k3, an indirect measure of choline kinase activity, was calculated using a two site compartmental model as previously described (29, 30). C, Ratio of betaine to phosphocholine in tumors. Metabolites were quantified by radio-HPLC at 15 and 60 min post injection of tracer. Mean values (n=4) and SEM are shown. * P<0.05; *** P<0.001. Abbreviations: p-choline, phosphocholine.



FIG. 25 shows dynamic uptake and metabolic stability of 18F-D4-choline in tumors of different histological origin. A, The tumor time versus radioactivity curve (TAC) obtained from 60 min dynamic PET imaging. Mean±SEM (n=3-5 mice per group). B, Metabolic profile of 18F-D4-choline in tumors. Radiolabelled metabolite profile in HCT116 tumor xenografts was assessed post PET imaging using radio-HPLC. Mean values (n=3) and SEM are shown. C, Choline kinase expression in malignant melanoma, prostate adenocarcinoma and colon carcinoma tumors. Representative western blot from tumor lysates (n=3 xenografts per tumor cell line). Actin was used as a loading control. Abbreviations: CKα, choline kinase alpha.



FIG. 26 shows effect of tumor size on 18F-D4-choline uptake and retention. Tracer uptake profiles were examined following 60 min dynamic PET imaging in PC3-M tumors at 100 mm3 () and 200 mm3 (◯). A, The tumor time versus radioactivity curve using average decay-corrected counts. Mean±SEM (n=3-5 mice per group). B, The tumor time versus radioactivity curve using the maximum voxel decay-corrected counts. Mean±SEM (n=3-5).



FIG. 27 shows analyte identification on radio-chromatograms. Representative radio-chromatograms of 18F-D4-choline-treated HCT116 cell lysates. A, 1 h uptake of 18F-D4-choline into HCT116 cells followed by cell lysis and 1 h incubation with vehicle at 37° C. B, 1 h uptake of 18F-D4-choline into HCT116 cells followed by cell lysis and 1 h incubation with alkaline phosphatase dissolved in vehicle. The labeled peaks are: 1, 18F-D4-choline; 2, 18F-D4-phosphocholine.



FIG. 28 shows choline oxidase treatment of 18F-D4-choline. A, Representative radio-chromatogram of 18F-D4-choline. B, 18F-D4-choline chromatogram following 20 min treatment with choline oxidase. C, 18F-D4-choline chromatogram following 40 min treatment. The labelled peaks are: 1, 18F-D4-betainealdehyde; 2, 18F-D4-betaine; 3, 18F-D4-choline.



FIG. 29 shows correlation between total kidney activity and % radioactivity retained as phosphocholine. Data were derived from 11C-choline, 11C-D4-choline and 18F-D4-choline uptake values and metabolism at 2, 15, 30 and 60 min post tracer injection.



FIG. 30 shows 11C-choline (◯), 11C-D4-choline (▴) and 18F-D4-choline (▪) PET imaging analysis in HCT116 tumors. The tumor time versus radioactivity curve (TAC) over the initial 14 min of the dynamic PET scans to illustrate subtle variations in tracer kinetics. Mean±SEM (n=4 mice per group).



FIG. 31 shows time course of 18F-D4-choline uptake in vitro in human melanoma (), prostate (▴) and colon (▪) cancer cell lines. Uptake was measured in vehicle-treated (closed symbols) and hemicholinium-3-treated cells (5 mM; open symbols). Mean values+SEM are shown (n=3). Insert: representative western blot of choline kinase-α expression in the three cell lines. Actin was used as a loading control. Abbreviations: CKα, choline kinase alpha.



FIG. 32 shows representative axial PET-CT images of PC3-M tumor-bearing mice (summed activity 30-60 min) at 100 mm3 and 200 mm3 respectively. Tumor margins, indicated from CT image, are outlined in red.





SUMMARY OF THE INVENTION

The present invention provides a compound of Formula (III):




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


C* is a radioisotope of carbon;


X, Y and Z are each independently hydrogen, deuterium (D), a halogen selected from F, Cl, Br, and I, alkyl, alkenyl, alkynl, aryl, heteroaryl, heterocyclyl group; and


Q is an anionic counterion; with the proviso the compound of Formula (III) is not 11C-choline.


DETAILED DESCRIPTION OF THE INVENTION

The present invention provides a novel radiolabeled choline analog compound of formula (I):




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


X and Y are each independently hydrogen, deuterium (D), or F;


Z is a halogen selected from F, Cl, Br, and I or a radioisotope; and


Q is an anionic counterion;


with the proviso that said compound of formula (I) is not fluoromethylcholine, fluoromethyl-ethyl-choline, fluoromethyl-propyl-choline, fluoromethyl-butyl-choline, fluoromethyl-pentyl-choline, fluoromethyl-isopropyl-choline, fluoromethyl-isobutyl-choline, fluoromethyl-sec-butyl-choline, fluoromethyl-diethyl-choline, fluoromethyl-diethanol-choline, fluoromethyl-benzyl-choline, fluoromethyl-triethanol-choline, 1,1-dideuterofluoromethylcholine, 1,1-dideuterofluoromethyl-ethyl-choline, 1,1-dideuterofluoromethyl-propyl-choline, or an [18F] analog thereof.


In a preferred embodiment of the invention, a compound of Formula (I) is provided wherein:


R1, R2, R3, and R4 are each independently hydrogen;


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


X and Y are each independently hydrogen, deuterium (D), or F;


Z is a halogen selected from F, Cl, Br, and I or a radioisotope;


Q is an anionic counterion;


with the proviso that said compound of formula (I) is not fluoromethylcholine, fluoromethyl-ethyl-choline, fluoromethyl-propyl-choline, fluoromethyl-butyl-choline, fluoromethyl-pentyl-choline, fluoromethyl-isopropyl-choline, fluoromethyl-isobutyl-choline, fluoromethyl-sec-butyl-choline, fluoromethyl-diethyl-choline, fluoromethyl-diethanol-choline, fluoromethyl-benzyl-choline, fluoromethyl-triethanol-choline, or an [18F] analog thereof.


In a preferred embodiment of the invention, a compound of Formula (I) is provided wherein:


R1 and R2 are each hydrogen;


R3 and R4 are each deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


X and Y are each independently hydrogen, deuterium (D), or F;


Z is a halogen selected from F, Cl, Br, and I or a radioisotope;


Q is an anionic counterion;


with the proviso that said compound of formula (I) is not 1,1-dideuterofluoromethylcholine, 1,1-dideuterofluoromethyl-ethyl-choline, 1,1-dideuterofluoromethyl-propyl-choline, or an [18F] analog thereof.


In a preferred embodiment of the invention, a compound of Formula (I) is provided wherein:


R1, R2, R3, and R4 are each deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


X and Y are each independently hydrogen, deuterium (D), or F;


Z is a halogen selected from F, Cl, Br, and I or a radioisotope;


Q is an anionic counterion.


According to the present invention, when Z of a compound of Formula (I) as described herein is a halogen, it can be a halogen selected from F, Cl, Br, and I; preferably, F.


According to the present invention, when Z of a compound of Formula (I) as described herein is a radioisotope (hereinafter referred to as a “radiolabeled compound of Formula (I)”), it can be any radioisotope known in the art. Preferably, Z is a radioisotope suitable for imaging (e.g., PET, SPECT). More preferably Z is a radioisotope suitable for PET imaging. Even more preferably, Z is 18F, 76Br, 123I, 124 or 125I. Even more preferably, Z is 18F.


According to the present invention, Q of a compound of Formula (I) as described herein can be any anionic counterion known in the art suitable for cationic ammonium compounds. Suitable examples of Q include anionic: bromide (Br), chloride (Cl), acetate (CH3CH2C(O)O), or tosylate (OTos). In a preferred embodiment of the invention, Q is bromide (Br) or tosylate (OTos). In a preferred embodiment of the invention, Q is chloride (Cl) or acetate (CH3CH2C(O)O). In a preferred embodiment of the invention, Q is chloride (Cl).


According the invention, a preferred embodiment of a compound of Formula (I) is the following compound of Formula (Ia):




embedded image


wherein:


R1, R2, R3, and R4 are each independently deuterium (D);


R5, R6, and R7 are each hydrogen;


X and Y are each independently hydrogen;


Z is 18F;


Q is Cl.


According to the invention, a preferred compound of Formula (Ia) is [18F]fluoromethyl-[1,2-2H4]-choline ([18F]-D4-FCH). [18F]-D4-FCH is a more metabolically stable fluorocholine (FCH) analog. [18F]-D4-FCH offers numerous advantages over the corresponding 18F-non-deuterated and/or 18F-di-deuterated analog. For example, [18F]-D4-FCH exhibits increased chemical and enzymatic oxidative stability relative to [18F]fluoromethylcholine. [18F]-D4-FCH has an improved in vivo profile (i.e., exhibits better availability for in vivo imaging) relative to dideuterofluorocholine, [18F]fluoromethyl-[1-2H2]choline, that is over and above what could be predicted by literature precedence and is, thus, unexpected. [18F]-D4-FCH exhibits improved stability and consequently will better enable late imaging of tumors after sufficient clearance of the radiotracer from systemic circulation. [18F]-D4-FCH also enhances the sensitivity of tumor imaging through increased availability of substrate. These advantages are discussed in further detail below.


The present invention further provides a precursor compound of Formula (II):




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4.


The present invention further provides a method of making a precursor compound of Formula (II).


The present invention provides a compound of Formula (III):




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5;


m is an integer from 1-4;


C* is a radioisotope of carbon;


X, Y and Z are each independently hydrogen, deuterium (D), a halogen selected from F, Cl, Br, and I, alkyl, alkenyl, alkynl, aryl, heteroaryl, heterocyclyl group; and


Q is an anionic counterion; with the proviso the compound of Formula (III) is not 11C-choline.


According to the invention, C* of the compound of Formula (III) can be any radioisotope of carbon. Suitable examples of C* include, but are not limited to, 11C, 13C, and 14C. Q is a described for the compound of Formula (I).


In a preferred embodiment of the invention, a compound of Formula (III) is provided wherein C* is 11C; X and Y are each hydrogen; and Z is F.


In a preferred embodiment of the invention, a compound of Formula (III) is provided wherein C* is 11C; X, Y and Z are each hydrogen H; R1, R2, R3, and R4 are each deuterium (D); and R5, R6, and R7 are each hydrogen (11C-[1,2-2H4]choline or “11C-D4-choline”.


Pharmaceutical or Radiopharmaceutical Composition

The present invention provides a pharmaceutical or radiopharmaceutical composition comprising a compound for Formula (I), including a compound of Formula (Ia), each as defined herein together with a pharmaceutically acceptable carrier, excipient, or biocompatible carrier. According to the invention when Z of a compound of Formula (I) or (Ia) is a radioisotope, the pharmaceutical composition is a radiopharmaceutical composition.


The present invention further provides a pharmaceutical or radiopharmaceutical composition comprising a compound for Formula (I), including a compound of Formula (Ia), each as defined herein together with a pharmaceutically acceptable carrier, excipient, or biocompatible carrier suitable for mammalian administration.


The present invention provides a pharmaceutical or radiopharmaceutical composition comprising a compound for Formula (III), as defined herein together with a pharmaceutically acceptable carrier, excipient, or biocompatible carrier.


The present invention further provides a pharmaceutical or radiopharmaceutical composition comprising a compound for Formula (III), as defined herein together with a pharmaceutically acceptable carrier, excipient, or biocompatible carrier suitable for mammalian administration.


As would be understood by one of skill in the art, the pharmaceutically acceptable carrier or excipient can be any pharmaceutically acceptable carrier or excipient known in the art.


The “biocompatible carrier” can be any fluid, especially a liquid, in which a compound of Formula (I), (Ia), or (III) can be suspended or dissolved, such that the pharmaceutical composition is physiologically tolerable, e.g., can be administered to the mammalian body without toxicity or undue discomfort. The biocompatible carrier is suitably an injectable carrier liquid such as sterile, pyrogen-free water for injection; an aqueous solution such as saline (which may advantageously be balanced so that the final product for injection is either isotonic or not hypotonic); an aqueous solution of one or more tonicity-adjusting substances (e.g., salts of plasma cations with biocompatible counterions), sugars (e.g., glucose or sucrose), sugar alcohols (e.g., sorbitol or mannitol), glycols (e.g., glycerol), or other non-ionic polyol materials (e.g., polyethyleneglycols, propylene glycols and the like). The biocompatible carrier may also comprise biocompatible organic solvents such as ethanol. Such organic solvents are useful to solubilise more lipophilic compounds or formulations. Preferably the biocompatible carrier is pyrogen-free water for injection, isotonic saline or an aqueous ethanol solution. The pH of the biocompatible carrier for intravenous injection is suitably in the range 4.0 to 10.5.


The pharmaceutical or radiopharmaceutical composition may be administered parenterally, i.e., by injection, and is most preferably an aqueous solution. Such a composition may optionally contain further ingredients such as buffers; pharmaceutically acceptable solubilisers (e.g., cyclodextrins or surfactants such as Pluronic, Tween or phospholipids); pharmaceutically acceptable stabilisers or antioxidants (such as ascorbic acid, gentisic acid orpara-aminobenzoic acid). Where a compound of Formula (I), (Ia), or (III) is provided as a radiopharmaceutical composition, the method for preparation of said compound may further comprise the steps required to obtain a radiopharmaceutical composition, e.g., removal of organic solvent, addition of a biocompatible buffer and any optional further ingredients. For parenteral administration, steps to ensure that the radiopharmaceutical composition is sterile and apyrogenic also need to be taken. Such steps are well-known to those of skill in the art.


Preparation of a Compound of the Invention

The present invention provides a method to prepare a compound for Formula (I), including a compound of Formula (Ia), wherein said method comprises reaction of the precursor compound of Formula (II) with a compound of Formula (IIIa) to form a compound of Formula (I) (Scheme A):




embedded image


wherein the compounds of Formulae (I) and (II) are each as described herein and the compound of Formula (IIIa) is as follows:





ZXYC-Lg  (IIIa)


wherein X, Y and Z are each as defined herein for a compound of Formula (I) and “Lg” is a leaving group. Suitable examples of “Lg” include, but are not limited to, bromine (Br) and tosylate (OTos). A compound of Formula (IIIa) can be prepared by any means known in the art including those described herein.


Synthesis of a compound of Formula (IIIa) wherein Z is F; X and Y are both H and the Lg is OTos (i.e., fluoromethyltosylate) can be achieved as set forth in Scheme 3 below:




embedded image


wherein: i: Silver p-toluenesulfonate, MeCN, reflux, 20 h;

    • ii: KF, MeCN, reflux, 1 h.


      According to Scheme 3 above:


(a) Synthesis of Methylene Ditosylate

Commercially available diiodomethane can be reacted with silver tosylate, using the method of Emmons and Ferris, to give methylene ditosylate (Emmons, W. D., et al., “Metathetical Reactions of Silver Salts in Solution. II. The Synthesis of Alkyl Sulfonates”, Journal of the American Chemical Society, 1953; 75:225).


(b) Synthesis of Cold Fluoromethyltosylate

Fluoromethyltosylate can be prepared by nucleophilic substitution of Methylene ditosylate from step (a) using potassium fluoride/Kryptofix K222 in acetonitrile at 80° C. under standard conditions.


When Z is a radioisotope, the radioisotope can be introduced by any means known by one of skill in the art. For example, the radioisotope [18F]-fluoride ion (18F) is normally obtained as an aqueous solution from the nuclear reaction 18O(p,n)18F and is made reactive by the addition of a cationic counterion and the subsequent removal of water. Suitable cationic counterions should possess sufficient solubility within the anhydrous reaction solvent to maintain the solubility of 18F. Therefore, counterions that have been used include large but soft metal ions such as rubidium or caesium, potassium complexed with a cryptand such as Kryptofix™, or tetraalkylammonium salts. A preferred counterion is potassium complexed with a cryptand such as Kryptofix™ because of its good solubility in anhydrous solvents and enhanced 18F reactivity. 18F can also be introduced by nucleophilic displacement of a suitable leaving group such as a halogen or tosylate group. A more detailed discussion of well-known 18F labelling techniques can be found in Chapter 6 of the “Handbook of Radiopharmaceuticals” (2003; John Wiley and Sons: M. J. Welch and C. S. Redvanly, Eds.). For example, [18F]Fluoromethyltosylate can be prepared by nucleophilic substitution of Methylene ditosylate with [18F]-fluoride ion in acetonitrile containing 2-10% water (see Neal, T. R., et al., Journal of Labelled Compounds and Radiopharmaceuticals 2005; 48:557-68).


Automated Synthesis

In a preferred embodiment, the method to prepare a compound for Formula (I), including a compound of Formula (Ia), is automated. For example, [18F]-radiotracers may be conveniently prepared in an automated fashion by means of an automated radiosynthesis apparatus. There are several commercially-available examples of such platform apparatus, including TRACERlab™ (e.g., TRACERlab™ MX) and FASTlab™ (both from GE Healthcare Ltd.). Such apparatus commonly comprises a “cassette”, often disposable, in which the radiochemistry is performed, which is fitted to the apparatus in order to perform a radiosynthesis. The cassette normally includes fluid pathways, a reaction vessel, and ports for receiving reagent vials as well as any solid-phase extraction cartridges used in post-radiosynthetic clean up steps. Optionally, in a further embodiment of the invention, the automated radiosynthesis apparatus can be linked to a high performance liquid chromatograph (HPLC).


The present invention therefore provides a cassette for the automated synthesis of a compound of Formula (I), including a compound of Formula (Ia), each as defined herein comprising:

    • i) a vessel containing the precursor compound of Formula (II) as defined herein; and
      • a. means for eluting the contents of the vessel of step (i) with a compound of Formula (IIIa) as defined herein.


        For the cassette of the invention, the suitable and preferred embodiments of the precursor compound of Formulae (II) and (IIIa) are each as defined herein.


In one embodiment of the invention, a method of making a compound of Formula (I), including a compound of Formula (Ia), each as described herein, that is compatible with FASTlab™ from a protected ethanolamine precursor that requires no HPLC purification step is provided.


The radiosynthesis of [18F]fluoromethyl-[1,2-2H4]choline (18F-D4-FCH) can be performed according to the methods and examples described herein. The radiosynthesis of 18F-D4-FCH can also be performed using commercially available synthesis platforms including, but not limited to, GE FASTlab™ (commercially available from GE Healthcare Inc.).


An example of a FASTlab™ radiosynthetic process for the preparation of [18F]fluoromethyl-[1,2-2H4]choline from a protected precursor is shown in Scheme 5:




embedded image


wherein:


a. Preparation of [18F]KF/K222/K2CO3 complex as described in more detail below;


b. Preparation of [18F]FCH2OTs as described in more detail below;


c. SPE purification of [18F]FCH2OTs as described in more detail below;


d. Radiosynthesis of O-PMB-[18F]-D4-Choline (O-PMB-[18F]-D4-FCH) as described in more detail below; and


e. Purification & formulation of [18F]-D4-Choline (18F-D4-FCH) as the hydrochloric salt as described in more detail below.


The automation of [18F]fluoro-[1,2-2H4]choline or [18F]fluorocholine (from the protected precursor) involves an identical automated process (and are prepared from the fluoromethylation of O-PMB-N,N-dimethyl-[1,2-2H4]ethanolamine and O-PMB-N,N-dimethylethanolamine respectively).


According to one embodiment of the present invention, FASTlab™ syntheses of [18F]fluoromethyl-[1,2-2H4]choline or [18F]fluoromethylcholine comprises the following sequential steps:


(i) Trapping of [18F]fluoride onto QMA;


(ii) Elution of [18F]fluoride from a QMA;


(iii) Radiosynthesis of [18F]FCH2OTs;


(iv) SPE clean up of [18F]FCH2OTs;


(v) Reaction vessel clean up;


(vi) Drying reaction vessel and [18F]fluoromethyl tosylate retained on SPE t-C18 plus simultaneously;


(vii) Alkylation reaction;


(viii) Removal of unreacted O-PMB-precursor; and


(ix) Deprotection & formulation.


Each of steps (i)-(ix) are described in more detail below.


In one embodiment of the present invention, steps (i)-(ix) above are performed on a cassette as described herein. One embodiment of the present invention is a cassette capable of performing steps (i)-(ix) for use in an automated synthesis platform. One embodiment of the present invention is a cassette for the radiosynthesis of [18F]fluoromethyl-[1,2-2H4]choline ([18F]-D4-FCH) or [18F]fluoromethylcholine from a protected precursor. An example of a cassette of the present invention is shown in FIG. 5b.


(i) Trapping of [18F]Fluoride onto QMA


[18F]fluoride (typically in 0.5 to 5 mL H218O) is passed through a pre-conditioned Waters QMA cartridge.


(Ii) Elution of [18F]Fluoride from a QMA


The eluent, as described in Table 1 is withdrawn into a syringe from the eluent vial and passed over the Waters QMA into the reaction vessel. This procedure elutes [18F]fluoride into the reaction vessel. Water and acetonitrile are removed using a well-designed drying cycle of “nitrogen/vacuum/heating/cooling”.


(Iii) Radiosynthesis of [18F]FCH2OTs

Once the K[18F]Fluoride/K222/K2CO3 complex of (ii) is dry, CH2(OTs)2 methylene ditosylate in a solution containing acetonitrile and water is added to the reaction vessel containing the K[18F]fluoride/K222/K2CO3 complex. The resulting reaction mixture will be heated (typically to 110° C. for 10 min), then cooled down (typically to 70° C.).


(Iv) SPE Clean Up of [18F]FCH2OTs

Once radiosynthesis of [18F]FCH2OTs is completed and the reaction vessel is cooled, water is added into the reaction vessel to reduce the organic solvent content in the reaction vessel to approximately 25%. This diluted solution is transferred from the reaction vessel and through the t-C18-light and t-C18 plus cartridges—these cartridges are then rinsed with 12 to 15 mL of a 25% acetonitrile/75% water solution. At the end of this process:

    • the methylene ditosylate remains trapped on the t-C18-light and
    • the [18F]FCH2OTs, tosyl-[18F]fluoride remains trapped on the t-C18 plus.


(v) Reaction Vessel Clean Up

The reaction vessel was cleaned (using ethanol) prior to the alkylation of [18F]fluoroethyl tosylate and O-PMB-DMEA precursor.


(Vi) Drying Reaction Vessel and [18F]Fluoromethyl Tosylate Retained on SPE t-C18 Plus Simultaneously


Once clean up (v) was completed, the reaction vessel and the [18F]fluoromethyl tosylate retained on SPE t-C18 plus was dried simultaneously.


(Vii) Alkylation Reaction

Following step (vi), the [18F]FCH2OTs (along with tosyl-[18F]fluoride) retained on the t-C18 plus was eluted into the reaction vessel using a mixture of O-PMB-N,N-dimethyl-[1,2-2H4]ethanolamine (or O-PMB-N,N-dimethylethanolamine) in acetonitrile.


The alkylation of [18F]FCH2OTs with O-PMB-precursor was achieved by heating the reaction vessel (typically 110° C. for 15 min) to afford [18F]fluoro-[1,2-2H4]choline (or O-PMB-[18F]fluorocholine).


(Viii) Removal of Unreacted O-PMB-Precursor

Water (3 to 4 mL) was added to the reaction and this solution was then passed through a pre-treated CM cartridge, followed by an ethanol wash—typically 2×5 mL (this removes unreacted O-PMB-DMEA) leaving “purified” [18F]fluoro-[1,2-2H4]choline (or O-PMB-[18F]fluorocholine) trapped onto the CM cartridge.


(ix) Deprotection & Formulation

Hydrochloric acid was passed through the CM cartridge into a syringe: this resulted in the deprotection of O-PMB-[18F]fluorocholine (the syringe contains [18F]fluorocholine in a HCl solution). Sodium acetate was then added to this syringe to buffer to pH 5 to 8 affording [18F]-D4-choline (or [18F]choline) in an acetate buffer. This buffered solution is then transferred to a product vial containing a suitable buffer.


Table 1 provides a listing of reagents and other components required for preparation of [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) (or [18F]fluoromethylcholine) radiocassette of the present invention:










TABLE 1





Reagent/Component
Description







Eluents
Eluent contains either:



K222/K2CO3 water/acetonitrile or



K222/KHCO3 water/acetonitrile or



18-crown-6/K2CO3 water/acetonitrile or



18-crown-6/KHCO3 water/acetonitrile.


25% acetonitrile/75% water
5 mL acetonitrile/15 mL water.


Ethanol
35 mL of ethanol


CH2(OTs)2
methylene ditosylate in an aqueous



acetonitrile solution


t-C18 light
SPE cartridge commercially available from



Waters (Milford, MA, USA)



Preconditioned by passing acetonitrile and



water (2 mL each) through


CM light
Commercially available from Waters


cartridge
(Milford, MA, USA). Preconditioned by



passing through 1M hydrochloric acid and



water (2 mL each).


PMB-O-precursor
O-PMB-N,N-dimethyl-[1,2-




2H4]ethanolamine and O-PMB-N,N-




dimethylethanolamine in anhydrous



acetonitrile


HCl
hydrochloric acid [1 to 5M]


NaOAC
sodium acetate solution [1 to 5M]


Water bag
100 mL water


t-C18 plus
SPE cartridge commercially available from



Waters (Milford, MA, USA)



Preconditioned by passing acetonitrile and



water (2 mL each) through


Ion exchange cartridge
Water pre-conditioned QMA light carb



commercially available from Waters



(Milford, MA, USA)









According to one embodiment of the present invention, FASTlab™ synthesis of [18F]fluoromethyl-[1,2-2H4]choline via an unprotected precursor comprises the following sequential steps as depicted in Scheme 6 below:




embedded image


1. Recovery of [18F]fluoride from QMA;


2 Preparation of K[18F]F/K222/K2CO3 complex;


3 Radiosynthesis of 18FCH2OTs;


4 SPE cleanup of 18FCH2OTs;


5 Clean up of reaction vessel cassette and syringe;


6 Drying of reaction vessel and C18 SepPak;


7 Elution off and coupling of 18FCH2OTs with D4-DMEA;


8 Transfer of reaction mixture onto CM cartridge;


9 Clean up of cassette and syringe;


10 Washing of CM cartridge with dilute aq ammonia solution, Ethanol and water;


11 Elution of [18F]fluoromethyl-[1,2-2H4]choline from CM cartridge with 0.09% sodium chloride (5 ml), followed by water (5 ml).


In one embodiment of the present invention, steps (1)-(11) above are performed on a cassette as described herein. One embodiment of the present invention is a cassette capable of performing steps (1)-(11) for use in an automated synthesis platform. One embodiment of the present invention is a cassette for the radiosynthesis of [18F]fluoromethyl-[1,2-2H4]choline ([18F]-D4-FCH) from an unprotected precursor. An example of a cassette of the present invention is shown in FIG. 5a.


Table 2 provides a listing of reagents and other components required for preparation of [18F]fluoromethyl-[1,2-2H4]choline (D4-FCH) (or [18F]fluoromethylcholine) via an unprotected precursor radiocassette of the present invention:










TABLE 2





Reagent/Component
Description







Sep-Pak light QMA
Commercially available from Waters


Carbonate cartridge
(Milford, MA, USA). Used as supplied.


Eluent prepared from stock
K2CO3: 17.9 mg/ml in water: 200 ul.


solutions:
Kryptofix222: 12 mg/ml in acetonitrile:



800 ul.


Organic wash for C18
15% acetonitrile in water, preloaded into


Sep-Pak pair
vial.


Bulk ethanol
50 ml preloaded into vial


CH2(OTs)2
4.4 mg of methylene ditosylate dissolved



into 1.25 ml acetonitrile containing 2%



water. Solution pre-loaded into vial.


t-C18 Sep-Pak light
SPE cartridge commercially available



from Waters (Milford, MA, USA).



Preconditioned by passing acetonitrile



then water through.


t-C18 Sep-Pak Plus
SPE cartridge commercially available



from Waters (Milford, MA, USA).



Preconditioned by passing acetonitrile



then water through.


Deuterated
Custom synthesis. 150-200 ul dissolved


dimethylethanolamine
into 1.4 ml acetonitrile. Preloaded into



vial.


Water bag
100 ml bag of sterile purified water.


Aqueous ammonia solution
10-15 ul of concentrated (30%)



ammonia in 10 ml water. 4 ml of this



solution preloaded into vial.


Sep-Pak light CM cartridge
Cartridge commercially available from



Waters (Milford, MA, USA). Used as



supplied.


Sodium Chloride for product
0.09% sodium chloride solution prepared


formulation
from 0.9% sodium chloride BP and water



for injection. BP.









Imaging Method

The radiolabeled compound of the invention, as described herein, will be taken up into cells via cellular transporters or by diffusion. In cells where choline kinase is overexpressed or activated the radiolabeled compound of the invention, as described herein, will be phosphorylated and trapped within that cell. This will form the primary mechanism of detecting neoplastic tissue.


The present invention further provides a method of imaging comprising the step of administering a radiolabeled compound of the invention or a pharmaceutical composition comprising a radiolabeled compound of the invention, each as described herein, to a subject and detecting said radiolabeled compound of the invention in said subject. The present invention further provides a method of detecting neoplastic tissue in vivo using a radiolabeled compound of the invention or a pharmaceutical composition comprising a radiolabeled compound of the invention, each as described herein. Hence the present invention provides better tools for early detection and diagnosis, as well as improved prognostic strategies and methods to easily identify patients that will respond or not to available therapeutic treatments. As a result of the ability of a compound of the invention to detect neoplastic tissue, the present invention further provides a method of monitoring therapeutic response to treatment of a disease state associated with the neoplastic tissue.


In a preferred embodiment of the invention, the radiolabeled compound of the invention for use in a method of imaging of the invention, as described herein, is a radiolabeled compound of Formula (I).


In a preferred embodiment of the invention, the radiolabeled compound of the invention for use in a method of imaging of the invention, as described herein, is a radiolabeled compound of Formula (III).


As would be understood by one of skill in the art the type of imaging (e.g., PET, SPECT) will be determined by the nature of the radioisotope. For example, if the radiolabeled compound of Formula (I) contains 18F it will be suitable for PET imaging.


Thus the invention provides a method of detecting neoplastic tissue in vivo comprising the steps of:

    • i) administering to a subject a radiolabeled compound of the invention or a pharmaceutical composition comprising a radiolabeled compound of the invention, each as defined herein;
    • ii) allowing said a radiolabeled compound of the invention to bind neoplastic tissue in said subject;
    • iii) detecting signals emitted by said radioisotope in said bound radiolabeled compound of the invention;
    • iv) generating an image representative of the location and/or amount of said signals; and,
    • v) determining the distribution and extent of said neoplastic tissue in said subject.


The step of “administering” a radiolabeled compound of the invention is preferably carried out parenterally, and most preferably intravenously. The intravenous route represents the most efficient way to deliver the compound throughout the body of the subject. Intravenous administration neither represents a substantial physical intervention nor a substantial health risk to the subject. The radiolabeled compound of the invention is preferably administered as the radiopharmaceutical composition of the invention, as defined herein. The administration step is not required for a complete definition of the imaging method of the invention. As such, the imaging method of the invention can also be understood as comprising the above-defined steps (ii)-(v) carried out on a subject to whom a radiolabeled compound of the invention has been pre-administered.


Following the administering step and preceding the detecting step, the radiolabeled compound of the invention is allowed to bind to the neoplastic tissue. For example, when the subject is an intact mammal, the radiolabeled compound of the invention will dynamically move through the mammal's body, coming into contact with various tissues therein. Once the radiolabeled compound of the invention comes into contact with the neoplastic tissue it will bind to the neoplastic tissue.


The “detecting” step of the method of the invention involves detection of signals emitted by the radioisotope comprised in the radiolabeled compound of the invention by means of a detector sensitive to said signals, e.g., a PET camera. This detection step can also be understood as the acquisition of signal data.


The “generating” step of the method of the invention is carried out by a computer which applies a reconstruction algorithm to the acquired signal data to yield a dataset. This dataset is then manipulated to generate images showing the location and/or amount of signals emitted by the radioisotope. The signals emitted directly correlate with the amount of enzyme or neoplastic tissue such that the “determining” step can be made by evaluating the generated image.


The “subject” of the invention can be any human or animal subject. Preferably the subject of the invention is a mammal. Most preferably, said subject is an intact mammalian body in vivo. In an especially preferred embodiment, the subject of the invention is a human.


The “disease state associated with the neoplastic tissue” can be any disease state that results from the presence of neoplastic tissue. Examples of such disease states include, but are not limited to, tumors, cancer (e.g., prostate, breast, lung, ovarian, pancreatic, brain and colon). In a preferred embodiment of the invention the disease state associated with the neoplastic tissue is brain, breast, lung, espophageal, prostate, or pancreatic cancer.


As would be understood by one of skill in the art, the “treatment” will be depend on the disease state associated with the neoplastic tissue. For example, when the disease state associated with the neoplastic tissue is cancer, treatment can include, but is not limited to, surgery, chemotherapy and radiotherapy. Thus a method of the invention can be used to monitor the effectiveness of the treatment against the disease state associated with the neoplastic tissue.


Other than neoplasms, a radiolabeled compound of the invention may also be useful in liver disease, brain disorders, kidney disease and various diseases associated with proliferation of normal cells. A radiolabeled compound of the invention may also be useful for imaging inflammation; imaging of inflammatory processes including rheumatoid arthritis and knee synovitis, and imaging of cardiovascular disease including artherosclerotic plaque.


Precursor Compound

The present invention provides a precursor compound of Formula (II):




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4.


In a preferred embodiment of the invention, a compound of Formula (II) is provided wherein:


R1, R2, R3, and R4 are each independently hydrogen;


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, or —CD(R8)2;


R8 is hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4.


In a preferred embodiment of the invention, a compound of Formula (II) is provided wherein:


R1 and R2 are each hydrogen;


R3 and R4 are each deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, or —CD(R8)2;


R8 is hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4.


In a preferred embodiment of the invention, a compound of Formula (II) is provided wherein:


R1, R2, R3, and R4 are each deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, or —CD(R8)2;


R8 is hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4.


According to the invention, compound of Formula (II) is a compound of Formula (IIa):




embedded image


In one embodiment of the invention, a compound of Formula (IIb) is provided:




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4; and


Pg is a hydroxyl protecting group.


In a preferred embodiment of the invention, a compound of Formula (IIb) is provided wherein Pg is a p-methoxybenyzl (PMB), trimethylsilyl (TMS), or a dimethoxytrityl (DMTr) group.


In a preferred embodiment of the invention, a compound of Formula (IIb) is provided wherein Pg is a p-methoxybenyzl (PMB) group.


In one embodiment of the invention, a compound of Formula (IIc) is provided:




embedded image


wherein:


R1, R2, R3, and R4 are each independently hydrogen or deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, —CH(R8)2, or —CD(R8)2;


R8 is independently hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4;


with the proviso that when R1, R2, R3, and R4 are each hydrogen, R5, R6, and R7 are each not hydrogen; and with the proviso that when R1, R2, R3, and R4 are each deuterium, R5, R6, and R7 are each not hydrogen.


In a preferred embodiment of the invention, a compound of Formula (IIc) is provided wherein:


R1, R2, R3, and R4 are each independently hydrogen;


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, or —CD(R8)2;


R8 is hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4; with the proviso that R5, R6, and R7 are each not hydrogen.


In a preferred embodiment of the invention, a compound of Formula (IIc) is provided wherein:


R1, R2, R3, and R4 are each deuterium (D);


R5, R6, and R7 are each independently hydrogen, R8, —(CH2)mR8, —(CD2)mR8, —(CF2)mR8, or —CD(R8)2;


R8 is hydrogen, —OH, —CH3, —CF3, —CH2OH, —CH2F, —CH2Cl, —CH2Br, —CH2I, —CD3, —CD2OH, —CD2F, CD2Cl, CD2Br, CD2I, or —C6H5; and


m is an integer from 1-4; with the proviso that R5, R6, and R7 are each not hydrogen.


In a preferred embodiment of the invention, a compound of Formula (IIc) is provided wherein:


R1 and R2 are each hydrogen; and


R3 and R4 are each deuterium (D).


A precursor compound of Formula (II), including a compound of Formula (IIa), (IIb) and (IIc), can be prepared by any means known in the art including those described herein. For example, the compound of Formula (IIa) can be synthesized by alkylation of dimethylamine in THF with 2-bromoethanol-1,1,2,2-d4 in the presence of potassium carbonate as shown in Scheme 1 below:




embedded image


wherein i=K2CO3, THF, 50° C., 19 h. The desired tetra-deuterated product can be purified by distillation. The 1H NMR spectrum of the compound of Formula (IIa) (FIG. 3) in deuteriochloroform showed only the peaks associated with the N,N-dimethyl groups and the hydroxyl of the alcohol; no peaks associated with the hydrogens of the methylene groups of the ethyl alcohol chain were observed. Consistent with this, the 13C NMR spectrum (FIG. 3) showed the large singlet associated with the N,N-dimethyl carbons; however, the peaks for the ethyl alcohol methylene carbons at 60.4 ppm and 62.5 ppm were substantially reduced in magnitude, suggesting the absence of the signal enhancement associated with the presence of a covalent carbon-hydrogen bond. In addition, the methylene peaks are both split into multiplets, indicating spin-spin coupling. Since 13C NMR is typically run with 1H decoupling, the observed multiplicity must be the result of carbon-deuterium bonding. On the basis of the above observations the isotopic purity of the desired product is considered to be >98% in favour of the 2H isotope (relative to the 1H isotope).


A di-deuterated analog of a precursor compound of Formula (II) can be synthesized from N,N-dimethylglycine via lithium aluminium hydride reduction as shown in Scheme 2 below:




embedded image


wherein i=LiAlD4, THF, 65° C., 24 h. 13C NMR analysis indicated that isotopic purity of greater than 95% in favor of the 2H isomer (relative to the 1H isotope) can be achieved.


According to the invention, the hydroxyl group of a compound of Formula (II), including a compound of Formula (IIa) can be further protected with a protecting group to give a compound of Formula (IIb):




embedded image


wherein Pg is any hydroxyl protecting group known in the art. Preferably, Pg is any acid labile hydroxyl protecting group including, for example, those described in “Protective Groups in Organic Synthesis”, 3rd Edition, A Wiley Interscience Publication, John Wiley & Sons Inc., Theodora W. Greene and Peter G. M. Wuts, pp 17-200. Preferably, Pg is a p-methoxybenzyl (PMB), trimethylsilyl (TMS), or a dimethoxytrityl (DMTr) group. More preferably, Pg is a p-methoxybenyzl (PMB) group.


Validation of [18F]Fluoromethyl-[1,2-2H4]Choline (D4-FCH)

Stability to oxidation resulting from isotopic substitution was evaluated in in vitro chemical and enzymatic models using [18F]fluoromethylcholine as standard. [18F]Fluoromethyl-[1,2-2H4]choline was then evaluated in in vivo models and compared to [11C]choline, [18F]fluoromethylcholine and [18F]Fluoromethyl-[1-2H2]choline:




embedded image


Potassium Permanganate Oxidation Study

The effect of deuterium substitution on bond strength was initially tested by evaluation of the chemical oxidation pattern of [18F]fluoromethylcholine and [18F]Fluoromethyl-[1,2-2H4]choline using potassium permanganate. Scheme 6 below details the base catalyzed potassium permanganate oxidation of [18F]fluoromethylcholine and [18F]Fluoromethyl-[1,2-2H4]choline at room temperature, with aliquots removed and analyzed by radio-HPLC at pre-selected time points:




embedded image


Reagents and Conditions: i) KMnO4, Na2CO3, H2O, rt.

The results are summarized in FIGS. 6 and 7. The radio-HPLC chromatogram (FIG. 6) showed a greater proportion of the parent compound remaining at 20 min for [18F]Fluoromethyl-[1,2-2H4]choline. The graph in FIG. 7 further showed a significant isotope effect for the deuterated analogue, [18F]Fluoromethyl-[1,2-2H4]choline, with nearly 80% of parent compound still present 1 hour post-treatment with potassium permanganate, compared to less than 40% of parent compound [18F]Fluoromethylcholine still present at the same time point.


Choline Oxidase Model

[18F]fluoromethylcholine and [18F]fluoromethyl-[1,2-2H4]choline were evaluated in a choline oxidase model (Roivainen, A., et al., European Journal of Nuclear Medicine 2000; 27:25-32). The graphical representation in FIG. 8 clearly shows that, in the enzymatic oxidative model, the deuterated compound is significantly more stable than the corresponding non-deuterated compound. At the 60 minute time point the radio-HPLC distribution of choline species revealed that for [18F]fluoromethylcholine the parent radiotracer was present at the level of 11±8%; at 60 minutes the corresponding parent deuterated radiotracer [18F]fluoromethyl-[1,2-2H4]choline was present at 29±4%. Relevant radio-HPLC chromatograms are shown in FIG. 9 and further exemplify the increased oxidative stability of [18F]fluoromethyl-[1,2-2H4]-choline relative to [18F]fluoromethylcholine. These radio-HPLC chromatograms contain a third peak, marked as ‘unknown’, that is speculated to be the intermediate oxidation product, betaine aldehyde.


In Vivo Stability Analysis

[18F]fluoromethyl-[1,2-2H4]-choline is more resistant to oxidation in vivo. The relative rates of oxidation of the two isotopically radiolabeled choline species, [18F]fluoromethylcholine and [18F]fluoromethyl-[1,2-2H4]-choline to their respective metabolites, [18F]fluoromethylcholine-betaine ([18F]-FCH-betaine) and [18F]fluoromethyl-[1,2-2H4]-choline-betaine ([18F]-D4-FCH-betaine) was evaluated by high performance liquid chromatography (HPLC) in mouse plasma after intravenous (i.v.) administration of the radiotracers. [18F]fluoromethyl-[1,2-2H4]-choline was found to be markedly more stable to oxidation than [18F]fluoromethylcholine. As shown in FIG. 10, [18F]fluoromethyl-[1,2-2H4]-choline was markedly more stable than [18F]fluoromethylcholine with ˜40% conversion of [18F]fluoromethyl-[1,2-2H4]-choline to [18F]-D4-FCH-betaine at 15 min after i.v. injection into mice compared to ˜80% conversion of [18F]fluoromethylcholine to [18F]-FCH-betaine. The time course for in vivo oxidation is shown in FIG. 10 showing overall improved stability of [18F]fluoromethyl-[1,2-2H4]-choline over [18F]fluoromethylcholine.


Biodistribution
Time Course Biodistribution

Time course biodistribution was carried out for [18F]fluoromethylcholine, [18F]fluoromethyl-[1-2H2]choline and [18F]fluoromethyl-[1,2-2H4]choline in nude mice bearing HCT116 human colon xenografts. Tissues were collected at 2, 30 and 60 minutes post-injection and the data summarized in FIG. 11A-C. The uptake values for [18F]fluoromethylcholine were in broad agreement with earlier studies (DeGrado, T. R., et al., “Synthesis and Evaluation of 18F-labeled Choline as an Oncologic Tracer for Positron Emisson Tomography: Initial Findings in Prostate Cancer”, Cancer Research 2000; 61:110-7). Comparison of the uptake profiles revealed a reduced uptake of radiotracer in the heart, lung and liver for the deuterated compounds [18F]fluoromethyl-[1-2H2]-choline and [18F]fluoromethyl-[1,2-2H4]-choline. The tumor uptake profile for the three radiotracers is shown in FIG. 11D and shows increased localization of radiotracer for the deuterated compounds relative to [18F]fluoromethylcholine at all time points. A pronounced increase in tumor uptake of [18F]fluoromethyl-[1,2-2H4]choline at the later time points is evident.


Distribution of Choline Metabolites

Metabolite analysis of tissues including liver, kidney and tumor by HPLC was also accomplished. Typical HPLC chromatograms of [18F]FCH and [18F]D4-FCH and their respective metabolites in tissues are shown in FIG. 12. Tumor distribution of metabolites was analyzed in a similar fashion (FIG. 13). Choline and its metabolites lack any UV chromophore to permit presentation of chromatograms of the cold unlabelled compound simultaneously with the radioactivity chromatograms. Thus, the presence of metabolites was validated by other chemical and biological means. Of note the same chromatographic conditions were used for characterization of the metabolites and retention times were similar. The identity of the phosphocholine peak was confirmed biochemically by incubation of the putative phosphocholine formed in untreated HCT116 tumor cells with alkaline phosphatase (FIG. 14). A high proportion of liver radioactivity was present as phosphocholine at 30 min post injection for both [18F]FCH and [18F]D4-FCH (FIG. 12). An unknown metabolite (possibly the aldehyde intermediate) was observed in both the liver (7.4±2.3%) and kidney (8.8±0.2%) samples of [18F]D4-FCH treated mice. In contrast, this unknown metabolite was not found in liver samples of [18F]FCH treated mice and only to a smaller extent (3.3±0.6%) in kidney samples. Notably 60.6±3.7% of [18F]D4-FCH derived kidney radioactivity was phosphocholine compared to 31.8±9.8% from [18F]FCH(P=0.03). Conversely, most of the [18F]FCH-derived radioactivity in the kidney was in the form of [18F]FCH-betaine; 53.5±5.3% compared to 20.6±6.2% for [18F]D4-FCH (FIG. 12). It could be argued that levels of betaine in plasma reflected levels in tissues such as liver and kidneys. Tumors showed a different HPLC profile compared to liver and kidneys; typical radio-HPLC chromatograms obtained from the analysis of tumor samples (30 min after intravenous injection of [18F]FCH, [18F]D4-FCH and [11C]choline) are shown in FIG. 12. In tumors, radioactivity was mainly in the form of phosphocholine in the case of [18F]D4-FCH (FIG. 13). In contrast [18F]FCH showed significant levels of [18F]FCH-betaine. In the context of late imaging, these results indicate that [18F]D4-FCH will be the superior radiotracer for PET imaging with an uptake profile that is easier to interpret.


The suitable and preferred aspects of any feature present in multiple aspects of the present invention are as defined for said features in the first aspect in which they are described herein. The invention is now illustrated by a series of non-limiting examples.


Isotopic Carbon Choline Analogs

The present invention provides a compound of Formula (III) as described herein. Such compounds are useful as PET imaging agents for tumor imaging, as described herein. In particular, a compound of Formula (III), as described herein, may not be excreted in the urine and hence provide more specific imaging of pelvic malignancies such as prostate cancer.


The present invention provides a method to prepare a compound for Formula (III), wherein said method comprises reaction of the precursor compound of Formula (II) with a compound of Formula (IV) to form a compound of Formula (III) (Scheme A):




embedded image


wherein the compounds of Formulae (I) and (III) are each as described herein and the compound of Formula (IV) is as follows:





ZXYC*-Lg  (IV)


wherein C*, X, Y and Z are each as defined herein for a compound of Formula (III) and “Lg” is a leaving group. Suitable examples of “Lg” include, but are not limited to, bromine (Br) and tosylate (OTos). A compound of Formula (IV) can be prepared by any means known in the art including those described herein (e.g., analogous to Examples 5 and 7).


EXAMPLES

Reagents and solvents were purchased from Sigma-Aldrich (Gillingham, UK) and used without further purification. Fluoromethylcholine chloride (reference standard) was purchased from ABCR Gmbh & Co. (Karlsruhe, Germany). Isotonic saline (0.9% w/v) was purchased from Hameln Pharmaceuticals (Gloucester, UK). NMR Spectra were obtained using either a Bruker Avance NMR machine operating at 400 MHz (1H NMR) and 100 MHz (13C NMR) or 600 MHz (1H NMR) and 150 MHz (13C NMR). Accurate mass spectroscopy was carried out on a Waters Micromass LCT Premier machine in positive electron ionisation (EI) or chemical ionisation (CI) mode. Distillation was carried out using a Bichi B-585 glass oven (Bichi, Switzerland).


Example 1
Preparation of N,N-dimethyl-[1,2-2H4]-ethanolamine (3)



embedded image


To a suspension of K2CO3 (10.50 g, 76 mmol) in dry THF (10 mL) was added dimethylamine (2.0 M in THF) (38 mL, 76 mmol) followed by 2-bromoethanol-1,1,2,2-d4 (4.90 g, 38 mmol) and the suspension heated to 50° C. under argon. After 19 h, thin layer chromatography (TLC) (ethyl acetate/alumina/I2) indicated complete conversion of (2) and the reaction mixture was allowed to cool to ambient temperature and filtered. Bulk solvent was then removed under reduced pressure. Distillation gave the desired product (3) as a colorless liquid, b.p. 78° C./88 mbar (1.93 g, 55%). 1H NMR (CDCl3, 400 MHz) δ 3.40 (s, 1H, OH), 2.24 (s, 6H, N(CH3)2). 13C NMR (CDCl3, 75 MHz) δ 62.6 (NCD2CD2OH), 60.4 (NCD2CD2OH), 47.7 (N(CH3)2). HRMS (EI)=93.1093 (M+). C4H72H4NO requires 93.1092.


Example 2
Preparation of N,N-dimethyl-[1-2H2]-ethanolamine (5)



embedded image


To a suspension of N,N-dimethylglycine (0.52 g, 5 mmol) in dry THF (10 mL) was added lithium aluminium deuteride (0.53 g, 12.5 mmol) and the resulting suspension refluxed under argon. After 24 h the suspension was allowed to cool to ambient temperature and poured onto sat. aq. Na2SO4 (15 mL) and adjusted to pH 8 with 1M Na2CO3, then washed with ether (3×10 mL) and dried (Na2SO4). Distillation gave the desired product (5) as a colorless liquid, b.p. 65° C./26 mbar (0.06 g, 13%). 1H NMR (CDCl3, 400 MHz) δ 2.43 (s, 2H, NCH2CD2), 2.25 (s, 6H, N(CH3)2), 1.43 (s, 1H, OH). 13C NMR (CDCl3, 150 MHz) δ 63.7 (NCH2CD2OH), 57.8 (NCH2CD2OH), 45.7 (N(CH3)2).


Example 3
Preparation of Fluoromethyltosylate (8)



embedded image


Methylene ditosylate (7) was prepared according to an established literature procedure and analytical data was consistent with reported values (Emmons, W. D., et al., Journal of the American Chemical Society, 1953; 75:2257; and Neal, T. R., et al., Journal of Labelled Compounds and Radiopharmaceuticals 2005; 48:557-68). To a solution of methylene ditosylate (7) (0.67 g, 1.89 mmol) in dry acetonitrile (10 mL) was added Kryptofix K222 [4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8.8.8]hexacosane] (1.00 g, 2.65 mmol) followed by potassium fluoride (0.16 g, 2.83 mmol). The suspension was then heated to 110° C. under nitrogen. After 1 h TLC (7:3 hexane/ethyl acetate/silica/UV254) indicated complete conversion of (7). The reaction mixture was diluted with ethyl acetate (25 mL), washed with water (2×15 mL) and dried over MgSO4. Chromatography (5→10% ethyl acetate/hexane) gave the desired product (8) as a colorless oil (40 mg, 11%). 1H NMR (CDCl3, 400 MHz) δ 7.86 (d, 2H, J=8 Hz, aryl CH), 7.39 (d, 2H, J=8 Hz, aryl CH), 5.77 (d, 1H, J=52 Hz, CH2F), 2.49 (s, 3H, tolyl CH3). 13C NMR (CDCl3) δ 145.6 (aryl), 133.8 (aryl), 129.9 (aryl), 127.9 (aryl), 98.1 (d, J=229 Hz, CH2F), 21.7 (tolyl CH3). HRMS (CI)=222.0604 (M+NH4)+. Calcd. for C8H13FNO3S 222.0600.


Example 4
Preparation of N,N-Dimethylethanolamine(O-4-methoxybenzyl)ether (O-PMB-DMEA)



embedded image


To a dry flask was added dimethylethanolamine (4.46 g, 50 mmol) and dry DMF (50 mL). The solution was stirred under argon and cooled in an ice bath. Sodium hydride (2.0 g, 50 mmol) was then added portionwise over 10 min and the reaction mixture then allowed to warm to room temperature. After 30 min 4-methoxybenzyl chloride (3.92 g, 25 mmol) was added dropwise over 10 min and the resulting mixture left to stir under argon. After 60 h GC-MS indicated reaction completion (disappearance of 4-methoxybenzyl chloride) and the reaction mixture was poured onto 1M sodium hydroxide (100 mL) and extracted with dichloromethane (DCM) (3×30 mL) then dried (Na2SO4). Column chromatography (0→10% methanol/DCM; neutral silica) gave the desired product (O-PMB-DMEA) as a yellow oil (1.46 g, 28%). 1H NMR (CDCl3, 400 MHz) δ 7.28 (d, 2H, J=8.6 Hz, aryl CH), 6.89 (d, 2H, J=8.6 Hz, aryl CH), 4.49 (s, 2H, —CH2—), 3.81 (s, 3H, OCH3), 3.54 (t, 2H, J=5.8, NCH2CH2O), 2.54 (t, 2H, J=5.8, NCH2CH2O), 2.28 (s, 6H, N(CH3)2). HRMS (ES)=210.1497 (M+H+). C12H20NO2 requires 210.1494.


Example 4a
Preparation of Dueterated Analogues of N,N-Dimethylethanolamine(O-4-methoxybenzyl)ether (O-PMB-DMEA)

The di- and tetra-deuterated analogs of N,N-Dimethylethanolamine(O-4-methoxybenzyl)ether can be prepared according to Example 4 from the appropriate di- or tetra-deuterated dimethylethanolamine.


Example 5
Preparation of Synthesis of [18F]fluoromethyl tosylate (9)



embedded image


To a Wheaton vial containing a mixture of K2CO3 (0.5 mg, 3.6 μmol, dissolved in 100 μL water), 18-crown-6 (10.3 mg, 39 μmol) and acetonitrile (500 μL) was added [18F]fluoride (˜20 mCi in 100 μL water). The solvent was then removed at 110° C. under a stream of nitrogen (100 mL/min). Afterwards, acetonitrile (500 μL) was added and distillation to dryness continued. This procedure was repeated twice. A solution of methylene ditosylate (7) (6.4 mg, 18 μmol) in acetonitrile (250 μL) containing 3% water was then added at ambient temperature followed by heating at 100° C. for 10-15 min., with monitoring by analytical radio-HPLC. The reaction was quenched by addition of 1:1 acetonitrile/water (1.3 mL) and purified by semi-preparative radio-HPLC. The fraction of eluent containing [18F]fluoromethyl tosylate (9) was collected and diluted to a final volume of 20 mL with water, then immobilized on a Sep Pak C18 light cartridge (Waters, Milford, Mass., USA) (pre-conditioned with DMF (5 mL) and water (10 mL)). The cartridge was washed with further water (5 mL) and then the cartridge, with [18F]fluoromethyl tosylate (9) retained, was dried in a stream of nitrogen for 20 min. A typical HPLC reaction profile for synthesis of [18F](13) is shown in FIG. 4A/4B below.


Example 6
Radiosynthesis of [18F]fluoromethylcholine Derivatives by Reaction with [18F]fluorobromomethane



embedded image


[18F]Fluorobromomethane (prepared according to Bergman et al (Appl Radiat Isot 2001; 54(6):927-33)) was added to a Wheaton vial containing the amine precursor N,N-dimethylethanolamine (150 μL) or N,N-dimethyl-[1,2-2H4]ethanolamine (3) (150 μL) in dry acetonitrile (1 mL), pre-cooled to 0° C. The vial was sealed and then heated to 100° C. for 10 min. Bulk solvent was then removed under a stream of nitrogen, then the sample remaining was redissolved in 5% ethanol in water (10 mL) and immobilized on a Sep-Pak CM light cartridge (Waters, Milford, Mass., USA) (pre-conditioned with 2 M HCl (5 mL) and water (10 mL)) to effect the chloride anion exchange. The cartridge was then washed with ethanol (10 mL) and water (10 mL) followed by elution of the radiotracer (11a) or (11c) using saline (0.5-2.0 mL) and passing through a sterile filter (0.2 μm) (Sartorius, Goettingen, Germany).


Example 7
Radiosynthesis of [18F]Fluoromethylcholine, [18F]fluoromethyl-[1-2H2]choline and [18F]fluoromethyl-[1,2-2H4]choline by Reaction with [18F]fluoromethylmethyl tosylate



embedded image


[18F]Fluoromethyl tosylate (9) (prepared according to Example 5) and eluted from the Sep-Pak cartridge using dry DMF (300 μL), was added in to a Wheaton vial containing one of the following precursors: N,N-dimethylethanolamine (150 μL); N,N-dimethyl-[1,2-2H4]ethanolamine (3) (150 μL) (prepared according to Example 1); or N,N-dimethyl-[1-2H2]ethanolamine (5) (150 μL) (prepared according to Example 2), and heated to 100° C. with stirring. After 20 min the reaction was quenched with water (10 mL) and immobilized on a Sep Pak CM light cartridge (Waters) (pre-conditioned with 2M HCl (5 mL) and water (10 mL)) in order to effect the chloride anion exchange and then washed with ethanol (5 mL) and water (10 mL) followed by elution of the radiotracer [18F]Fluoromethylcholine (12a), [18F]fluoromethyl-[1-2H2]choline (12b) or [18F]fluoromethyl-[1,2-2H4]choline [18F](12c) with isotonic saline (0.5-1.0 mL).


Example 8
Synthesis of Cold Fluoromethyltosylate (15)



embedded image




    • i: Silver p-toluenesulfonate, MeCN, reflux, 20 h;

    • ii: KF, MeCN, reflux, 1 h.


      According to Scheme 3 above:





(a) Synthesis of Methylene Ditosylate (14)

Commercially available diiodomethane (13) (2.67 g, 10 mmol) was reacted with silver tosylate (6.14 g, 22 mmol), using the method of Emmons and Ferris, to give methylene ditosylate (10) (0.99 g) in 28% yield (Emmons, W. D., et al., “Metathetical Reactions of Silver Salts in Solution. II. The Synthesis of Alkyl Sulfonates”, Journal of the American Chemical Society, 1953; 75:225).


(b) Synthesis of Cold Fluoromethyltosylate (15)

Fluoromethyltosylate (11) (0.04 g) was prepared by nucleophilic substitution of Methylene ditosylate (10) (0.67 g, 1.89 mmol) of Example 3(a) using potassium fluoride (0.16 g, 2.83 mmol)/Kryptofix K222 (1.0 g, 2.65 mmol) in acetonitrile (10 mL) at 80° C. to give the desired product in 11% yield.


Example 9
Synthesis of [18F]fluorobromomethane (17)



embedded image


Adapting the method of Bergman et al (Appl Radiat Isot 2001; 54(6):927-33), commercially available dibromomethane (16) is reacted with [18F]potassium fluoride/Kryptofix K222 in acetonitrile at 110° C. to give the desired [18F]fluorobromomethane (17), which is purified by gas-chromatography and trapped by elution into a pre-cooled vial containing acetonitrile and the relevant choline precursor.


Example 10
Analysis of Radiochemical Purity

Radiochemical purity for [18F]Fluoromethylcholine, [18F]fluoromethyl-[1-2H2]choline and [18F]fluoromethyl-[1,2-2H4]choline [18F] was confirmed by co-elution with a commercially available fluorocholine chloride standard. An Agilent 1100 series HPLC system equipped with an Agilent G1362A refractive index detector (RID) and a Bioscan Flowcount FC-3400 PIN diode detector was used. Chromatographic separation was performed on a Phenomenex Luna C18 reverse phase column (150 mm×4.6 mm) and a mobile phase comprising of 5 mM heptanesulfonic acid and acetonitrile (90:10 v/v) delivered at a flow rate of 1.0 mL/min.


Example 11
Enzymatic Oxidation Study Using Choline Oxidase

This method was adapted from that of Roivannen et al (Roivainen, A., et al., European Journal of Nuclear Medicine 2000; 27:25-32). An aliquot of either [18F]Fluoromethylcholine or [18F]fluoromethyl-[1,2-2H4]choline [18F](100 μL, ˜3.7 MBq) was added to a vial containing water (1.9 mL) to give a stock solution. Sodium phosphate buffer (0.1M, pH 7) (10 uL) containing choline oxidase (0.05 units/uL) was added to an aliquot of stock solution (190 uL) and the vial was then left to stand at room temperature, with occasional agitation. At selected time-points (5, 20, 40 and 60 minutes) the sample was diluted with HPLC mobile phase (buffer A, 1.1 mL), filtered (0.22 μm filter) and then ˜1 mL injected via a 1 mL sample loop onto the HPLC for analysis. Chromatographic separation was performed on a Waters C18 Bondapak (7.8×300 mm) column (Waters, Milford, Massachusetts, USA) at 3 mL/min with a mobile phase of buffer A, which contained acetonitrile, ethanol, acetic acid, 1.0 mol/L ammonium acetate, water, and 0.1 mol/L sodium phosphate (800:68:2:3:127:10 [v/v]) and buffer B, which contained the same constituents but in different proportions (400:68:44:88:400:10 [v/v]). The gradient program comprised 100% buffer A for 6 minutes, 0-100% buffer B for 10 minutes, 100-0% B in 2 minutes then 0% B for 2 minutes.


Example 12
Biodistribution

Human colon (HCT116) tumors were grown in male C3H-Hej mice (Harlan, Bicester, United Kingdom) as previously reported (Leyton, J., et al., Cancer Research 2005; 65(10):4202-10). Tumor dimensions were measured continuously using a caliper and tumor volumes were calculated by the equation: volume=(π/6)×a×b×c, where a, b, and c represent three orthogonal axes of the tumor. Mice were used when their tumors reached approximately 100 mm3. [18F]Fluoromethylcholine, [18F]fluoromethyl-[1-2H2]choline and [18F]fluoromethyl-[1,2-2H4]choline (˜3.7 MBq) were each injected via the tail vein into awake untreated tumor bearing mice. The mice were sacrificed at pre-determined time points (2, 30 and 60 min) after radiotracer injection under terminal anesthesia to obtain blood, plasma, tumor, heart, lung, liver, kidney and muscle. Tissue radioactivity was determined on a gamma counter (Cobra II Auto-Gamma counter, Packard Biosciences Co, Pangbourne, UK) and decay corrected. Data were expressed as percent injected dose per gram of tissue.


Example 13
Oxidation Potential of [18F]Fluoromethylcholine ([18F]FCH) and [18F]fluoromethyl-[1,2-2H4]choline ([18F]D4-FCH) In Vivo

[18F]FCH or [18F](D4-FCH) (80-100 μCi) was injected via the tail vein into anesthetized non-tumor bearing C3H-Hej mice; isofluorane/O2/N2O anesthesia was used. Plasma samples obtained at 2, 15, 30 and 60 minutes after injection were snap frozen in liquid nitrogen and stored at −80° C. For analysis, samples were thawed and kept at 4° C. To approximately 0.2 mL of plasma was added ice-cold acetonitrile (1.5 mL). The mixture was then centrifuged (3 minutes, 15,493×g; 4° C.). The supernatant was evaporated to dryness using a rotary evaporator (Heidoloph Instruments GMBH & CO, Schwabach, Germany) at a bath temperature of 45° C. The residue was suspended in mobile phase (1.1 mL), clarified (0.2 μm filter) and analyzed by HPLC. Liver samples were homogenized in ice-cold acetonitrile (1.5 mL) and then subsequently treated as per plasma samples. All samples were analyzed on an Agilent 1100 series HPLC system equipped with a γ-RAM Model 3 radio-detector (IN/US Systems inc., FL, USA). The analysis was based on the method of Roivannen (Roivainen, A., et al., European Journal of Nuclear Medicine 2000; 27:25-32) using a Phenomenex Luna SCX column (10μ, 250×4.6 mm) and a mobile phase comprising of 0.25 M sodium dihydrogen phosphate (pH 4.8) and acetonitrile (90:10 v/v) delivered at a flow rate of 2 ml/min.


Example 14
Distribution of Choline Metabolites

Liver, kidney, and tumor samples were obtained at 30 min. All samples were snap-frozen in liquid nitrogen. For analysis, samples were thawed and kept at 4° C. immediately before use. To ˜0.2 mL plasma was added ice-cold methanol (1.5 mL). The mixture was then centrifuged (3 min, 15,493×g, 4jC). The supernatant was evaporated to dryness using a rotary evaporator (Heidoloph Instruments) at a bath temperature of 40° C. The residue was suspended in mobile phase (1.1 mL), clarified (0.2 Am filter), and analyzed by HPLC. Liver, kidney, and tumor samples were homogenized in ice-cold methanol (1.5 mL) using an IKA Ultra-Turrax T-25 homogenizer and subsequently treated as per plasma samples (above). All samples were analyzed by radio-HPLC on an Agilent 1100 series HPLC system (Agilent Technologies) equipped with a γ-RAM Model 3 γ-detector (IN/US Systems) and Laura 3 software (Lablogic). The stationary phase comprised a Waters μBondapak C18 reverse-phase column (300×7.8 mm) (Waters, Milford, Mass., USA). Samples were analyzed using a mobile phase comprising solvent A (acetonitrile/water/ethanol/acetic acid/1.0 mol/L ammonium acetate/0.1 mol/L sodium phosphate; 800/127/68/2/3/10) and solvent B (acetonitrile/water/ethanol/acetic acid/1.0 mol/L ammonium acetate/0.1 mol/L sodiumphosphate; 400/400/68/44/88/10) with a gradient of 0% B for 6 min, then 0→100% B in 10 min, 100% B for 0.5 min, 100→0% B in 1.5 min then 0% B for 2 min, delivered at a flow rate of 3 mL/min.


Example 15
Metabolism of [18F]D4-FCH and [18F]FCH by HCT116 Tumor Cells

HCT116 cells were grown in T150 flasks in triplicate until they were 70% confluent and then treated with vehicle (1% DMSO in growth medium) or 1 μmol/L PD0325901 in vehicle for 24 h. Cells were pulsed for 1 h with 1.1 MBq of either [8F]D4—FCH or [18F]FCH. The cells were washed three times in ice-cold phosphate buffered saline (PBS), scraped into 5 mL PBS, and centrifuged at 500×g for 3 min and then resuspended in 2 mL ice-cold methanol for HPLC analysis as described above for tissue samples. To provide biochemical evidence that the 5′-phosphate was the peak identified on the HPLC chromatogram, cultured cells were treated with alkaline phosphatase as described previously (Barthel, H., et al., Cancer Res 2003; 63(13):3791-8). Briefly, HCT116 cells were grown in 100 mm dishes in triplicate and incubated with 5.0 MBq [18F]FCH for 60 min at 37° C. to form the putative [18F]FCH-phosphate. The cells were washed with 5 mL ice-cold PBS twice and then scraped and centrifuged at 750×g (4° C., 3 min) in 5 mL PBS. Cells were homogenized in 1 mL of 5 mmol/L Tris-HCl (pH 7.4) containing 50% (v/v) glycerol, 0.5 mmol/L MgCl2, and 0.5 mmol/L ZnCl2 and incubated with 10 units bacterial (type III) alkaline phosphatase (Sigma) at 37° C. in a shaking water bath for 30 min to dephosphorylate the [18F]FCH-phosphate. The reaction was terminated by adding ice-cold methanol. Samples were processed as per plasma above and analyzed by radio-HPLC. Control experiments were done without alkaline phosphatase.


Example 16
Small Animal PET Imaging

PET Imaging Studies.


Dynamic [18F]FCH and [18F]D4-FCH imaging scans were carried out on a dedicated small animal PET scanner, quad-HIDAC (Oxford Positron Systems). The features of this instrument have been described previously (Barthel, H., et al., Cancer Res 2003; 63(13):3791-8). For scanning the tail veins, vehicle- or drug-treated mice were cannulated after induction of anesthesia (isofluorane/O2/N2O). The animals were placed within a thermostatically controlled jig (calibrated to provide a rectal temperature of ˜37° C.) and positioned prone in the scanner. [18F]FCH or [18F]D4-FCH (2.96-3.7 MBq) was injected via the tail vein cannula and scanning commenced. Dynamic scans were acquired in list mode format over a 60 min period as reported previously (Leyton, J., et al., Cancer Research 2006; 66(15):7621-9). The acquired data were sorted into 0.5 mm sinogram bins and 19 time frames (0.5×0.5×0.5 mm voxels; 4×15, 4×60, and 11×300 s) for image reconstruction, which was done by filtered back-projection using a two-dimensional Hamming filter (cutoff 0.6). The image data sets were visualized using the Analyze software (version 6.0; Biomedical Imaging Resource, Mayo Clinic). Cumulative images of 30 to 60 min dynamic data were used for visualization of radiotracer uptake and to draw regions of interest. Regions of interest were defined manually on five adjacent tumor regions (each 0.5 mm thickness). Dynamic data from these slices were averaged for each tissue (liver, kidney, muscle, urine, and tumor) and at each of the 19 time points to obtain time versus radioactivity curves. Corresponding whole body time versus radioactivity curves representing injected radioactivity were obtained by adding together radioactivity in all 200×160×160 reconstructed voxels. Tumor radioactivity was normalized to whole-body radioactivity and expressed as percent injected dose per voxel (% ID/vox). The normalized uptake of radiotracer at 60 min (% ID/vox60) was used for subsequent comparisons. The average of the normalized maximum voxel intensity across five slices of tumor % IDvox60max was also use for comparison to account for tumor heterogeneity and existence of necrotic regions in tumor. The area under the curve was calculated as the integral of % ID/vox from 0 to 60 min.


Example 17
Effect of PD0325901 Treatment in Mice

Size-matched HCT116 tumor bearing mice were randomized to receive daily treatment by oral gavage of vehicle (0.5% hydroxypropyl methylcellulose+0.2% Tween 80) or 25 mg/kg (0.005 mL/g mouse) of the mitogenic extracellular kinase inhibitor, PD0325901, prepared in vehicle. [18F]D4-FCH-PET scanning was done after 10 daily treatments with the last dose administered 1 h before scanning. After imaging, tumors were snap-frozen in liquid nitrogen and stored at ˜80° C. for analysis of choline kinase A expression. The results are illustrated in FIGS. 18 and 19.


This exemplifies use of [18F]D4-FCH-PET as an early biomarker of drug response.


Most of the current drugs in development for cancer target key kinases involved in cell proliferation or survival. This example shows that in a xenograft model for which tumor shrinkage is not significant, growth factor receptor-Ras-MAP kinase pathway inhibition by the MEK inhibitor PD0325901 leads to a significant reduction in tumor [18F]D4-FCH uptake signifying inhibition of the pathway. The figure also shows that inhibition of [18F]D4-FCH uptake was due at least in part to the inhibition of choline kinase activity.


Example 18
Comparison of [18F]FCH and [18F]D4-FCH for Imaging

As illustrated in FIG. 16, [18F]FCH and [18F]D4-FCH were both rapidly taken up into tissues and retained. Tissue radioactivity increased in the following order: muscle<urine<kidney<liver. Given the predominance of phosphorylation over oxidation in the liver (FIG. 12), little differences were found in overall liver radioactivity levels between the two radiotracers. Liver radioactivity at levels 60 min after [18F]D4-FCH or [18F]FCH injection, % ID/vox60, was 20.92±4.24 and 18.75±4.28, respectively (FIG. 16). This is also in keeping with the lower levels betaine with [18F]D4-FCH injection than with [18F]FCH injection (FIG. 12). Thus, pharmacokinetics of the two radiotracers in liver determined by PET (which lacks chemical resolution) were similar. The lower kidney radioactivity levels for [18F]D4-FCH compared to [18F]FCH (FIG. 16), on the other hand, reflect the lower oxidation potential of [18F]D4-FCH in kidneys. The % ID/vox60 for [18F]FCH and [18F]D4-FCH were 15.97±4.65 and 7.59±3.91, respectively in kidneys (FIG. 16). Urinary excretion was similar between the radiotracers. Regions of interest (ROIs) that were drawn over the bladder showed % ID/vox60 values of 5.20±1.71 and 6.70±0.71 for [18F]D4-FCH and [18F]FCH, respectively. Urinary metabolites comprised mainly of the unmetabolized radiotracers. Muscle showed the lowest radiotracer levels of any tissue.


Despite the relatively high systemic stability of [18F]D4-FCH and high proportion of phosphocholine metabolites, higher tumor radiotracer uptake by PET in mice that were injected with [18F]D4-FCH compared to the [18F]FCH group was observed. FIG. 17 shows typical (0.5 mm) transverse PET image slices demonstrating accumulation of [18F]FCH and [18F]D4-FCH in human melanoma SKMEL-28 xenografts. In this mouse model, the tumor signal-to-background contrast was qualitatively superior in the [18F]D4-FCH PET images compared to [18F]FCH images. Both radiotracers had similar tumor kinetic profiles detected by PET (FIG. 17). The kinetics were characterized by rapid tumor influx with peak radioactivity at ˜1 min (FIG. 17). Tumor levels then equilibrated until ˜5 min followed by a plateau. The delivery and retention of [18F]D4-FCH were quantitatively higher than those for FCH (FIG. 17). The % ID/vox60 for [18F]D4-FCH and [18F]FCH were 7.43±0.47 and 5.50±0.49, respectively (P=0.04). Because tumors often present with heterogeneous population of cells, another imaging variable that is probably less sensitive to experimental noise was exploited—an average of the maximum pixel % ID/vox60 across 5 slices (% IDvox60max). This variable was also significantly higher for [18F]D4-FCH(P=0.05; FIG. 17). Furthermore, tumor area under the time versus radioactivity curve (AUC) was higher for D4-FCH mice than FCH(P=0.02). Although the 30 min time point was selected for a more detailed analysis of tissue samples, the percentage of parent compound in plasma was consistently higher for [18F]D4-FCH compared to [18F]FCH at earlier time points. Regarding imaging, tumor uptake for both radiotracers was similar at the early (15 min) and late (60 min) time points (Supplementary Table1). The earlier time points may be appropriate for pelvic imaging.


Example 19
Imaging Response to Treatment Having demonstrated that [18F]D4-FCH was a more stable fluorinated-choline analog for in vivo studies, the use of this radiotracer to measure response to therapy was investigated. These studies were performed in a reproducible tumor model system in which treatment outcomes had been previously characterized, i.e., the human colon carcinoma xenograft HCT116 treated with PD0325901 daily for 10 days (Leyton, J., et al., “Noninvasive imaging of cell proliferation following mitogenic extracellular kinase inhibition by PD0325901”, Mol Cancer Ther 2008; 7(9):3112-21). Drug treatment led to tumor stasis (reduction in tumor size by only 12.2% at day 10 compared to the pretreatment group); tumors of vehicle-treated mice increased by 375%. Tumor [18F]D4-FCH levels in PD0325901-treated mice peaked at approximately the same time as those of vehicle-treated ones, however, there was a marked reduction in radiotracer retention in the treated tumors (FIG. 18). All imaging variables decreased after 10 days of drug treatment (P=0.05, FIG. 18). This indicates that [18F]D4-FCH can be used to detect treatment response even under conditions where large changes in tumor size reduction are not seen (Leyton, J., et al., “Noninvasive imaging of cell proliferation following mitogenic extracellular kinase inhibition by PD0325901”, Mol Cancer Ther 2008; 7(9):3112-21). To understand the biomarker changes, the intrinsic cellular effect of PD0325901 on D4-FCH-phosphocholine formation was examined by treating exponentially growing HCT116 cells in culture with PD0325901 for 24 h and measuring the 60-min uptake of [18F]D4-FCH in vitro. As shown in FIG. 18, PD0325901 significantly inhibited [18F]D4-FCH-phosphocholine formation in drug-treated cells demonstrating that the effect of the drug in tumors is likely due to cellular effects on choline metabolism rather than hemodynamic effects.

To understand further the mechanisms regulating [18F]D4-FCH uptake with drug treatment, changes in CHKA expression in PD0325901 and vehicle-treated tumors excised after PET scanning were assessed. A significant reduction in CHKA protein expression was seen in vivo at day 10 (P=0.03) following PD0325901 treatment (FIG. 19) indicating that reduced CHKA expression contributed to the lower D[18F]-4-FCH uptake in drug-treated tumors. The drug-induced reduction of CHKA expression also occurred in vitro in exponentially growing cells treated with PD0325901.


Example 20
Statistics

Statistical analyses were done using the software GraphPad Prism version 4 (GraphPad). Between-group comparisons were made using the nonparametric Mann-Whitney test. Two-tailed P≦0.05 was considered significant.


Example 21

Materials and Methods


Cell Lines

HCT116 (LGC Standards, Teddington, Middlesex, UK) and PC3-M cells (donation from Dr Matthew Caley, Prostate Cancer Metastasis Team, Imperial College London, UK) were grown in RPMI 1640 media, supplemented with 10% fetal calf serum, 2 mM L-glutamine, 100 U·mL−1 penicillin and 100 μg·mL−1 streptomycin (Invitrogen, Paisley, Refrewshire, UK). A375 cells (donation from Professor Eyal Gottlieb, Beatson Institute for Cancer Research, Glasgow, UK) and were grown in high glucose (4.5 g/L) DMEM media, supplemented with 10% fetal calf serum, 2 mM L-glutamine, 100 U·mL−1 penicillin and 100 μg·mL−1 streptomycin (Invitrogen, Paisley, Refrewshire, UK). All cells were maintained at 37° C. in a humidified atmosphere containing 5% CO2.


Western Blots

Western blotting was performed using standard techniques. Cells were harvested and lysed in RIPA buffer (Thermo Fisher Scientific Inc., Rockford, Ill., USA). Membranes were probed using a rabbit anti-human choline kinase alpha polyclonal antibody (Sigma-Aldrich Co. Ltd, Poole, Dorset, UK; 1:500). A rabbit anti-actin antibody (Sigma-Aldrich Co. Ltd, Poole, Dorset, UK; 1:5000) was used as a loading control and a peroxidase-conjugated donkey anti-rabbit IgG antibody (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA; 1:2500) as the secondary antibody. Proteins were visualized using the Amersham ECL kit (GE Healthcare, Chalfont St Giles, Bucks, UK). Blots were scanned (Bio-Rad GS-800 Calibrated Densitometer; Bio-Rad, Hercules, Calif., USA) and signal quantification was performed by densitometry using scanning analysis software (Quantity One; Bio-Rad).


For analysis of tumor choline kinase expression, tumors at ˜100 mm3 were excised, placed in a Precellys 24 lysing kit 2 mL tube (Bertin Technoologies, Montigny-le-Bretonneux, France), containing 1.4 mm ceramic beads, and snap-frozen in liquid nitrogen. For homogenization, 1 mL of RIPA buffer was added to the lysing kit tubes which were homogenized in a Precellys 24 homogenizer (6500 RPM; 2×17 s with 20 s interval). Cell debris were removed by centrifugation prior to western blotting as described above.


In Vitro 18F-D4-choline Uptake


Cells (5×105) were plated into 6-well plates the night prior to analysis. On the day of the experiment, fresh growth medium, containing 40 μCi 18F-D4-choline, was added to individual wells. Cell uptake was measured following incubation at 37° C. in a humidified atmosphere of 5% CO2 for 60 min. Plates were subsequently placed on ice, washed 3 times with ice-cold PBS and lysed in RIPA buffer (Thermo Fisher Scientific Inc., Rockford, Ill., USA; 1 mL, 10 min). Cell lysate was transferred to counting tubes and decay-corrected radioactivity was determined on a gamma counter (Cobra II Auto-Gamma counter, Packard Biosciences Co, Pangbourne, UK). Aliquots were snap-frozen and used for protein determination following radioactive decay using a BCA 96-well plate assay (Thermo Fisher Scientific Inc., Rockford, Ill., USA). Data were expressed as percent of total radioactivity per mg protein. For hemicholinium-3 treatment (5 mM; Sigma-Aldrich), cells were incubated with the compound 30 min prior to addition of radioactivity and for the duration of the uptake time course.


In Vivo Tumor Models

All animal experiments were performed by licensed investigators in accordance with the United Kingdom Home Office Guidance on the Operation of the Animal (Scientific Procedures) Act 1986 and within the newly-published guidelines for the welfare and use of animals in cancer research (Workman P, Aboagye E O, Balkwill F, et al. Guidelines for the welfare and use of animals in cancer research. Br J Cancer. 2010; 102:1555-1577). Male BALB/c nude mice (aged 6-8 weeks; Charles River, Wilmington, Mass., USA) were used. Tumor cells (2×106) were injected subcutaneously on the back of mice and animals were used when the xenografts reached ˜100 mm3. Tumor dimensions were measured continuously using a caliper and tumor volumes were calculated by the equation: volume=(π/6)×a×b×c, where a, b, and c represent three orthogonal axes of the tumor.


In Vivo Tracer Metabolism

Radiolabeled metabolites from plasma and tissues were quantified using a method adapted from Smith G, Zhao Y, Leyton J, et al. Radiosynthesis and pre-clinical evaluation of [(18)F]fluoro-[1,2-(2)H(4)]choline. Nucl Med Biol. 2011; 38:39-51. Briefly, tumor-bearing mice under terminal anaesthesia were administered a bolus i.v. injection of one of the following radiotracers: 11C-choline, 11C-D4-choline (˜18.5 MBq) or 18F-D4-choline (˜3.7 MBq), and sacrificed by exsanguination via cardiac puncture at 2, 15, 30 or 60 min post radiotracer injection. For automated radiosynthesis methodology, see Example 22. Tumor, kidney and liver samples were immediately snap-frozen in liquid nitrogen. Aliquots of heparinized blood were rapidly centrifuged (14000 g, 5 min, 4° C.) to obtain plasma. Plasma samples were subsequently snap-frozen in liquid nitrogen and kept on dry ice prior to analysis.


For analysis, samples were thawed and kept at 4° C. immediately before use. To ice cold plasma (200 μl) was added ice cold methanol (1.5 mL) and the resulting suspension centrifuged (14000 g; 4° C.; 3 min). The supernatant was then decanted and evaporated to dryness on a rotary evaporator (bath temperature, 40° C.), then resuspended in HPLC mobile phase (Solvent A: acetonitrile/water/ethanol/acetic acid/1.0 M ammonium acetate/0.1 M sodium phosphate [800/127/68/2/3/10]; 1.1 mL). Samples were filtered through a hydrophilic syringe filter (0.2 μm filter; Millex PTFE filter, Millipore, Mass., USA) and the sample (˜1 mL) then injected via a 1 mL sample loop onto the HPLC for analysis. Tissues were homogenized in ice-cold methanol (1.5 mL) using an Ultra-Turrax T-25 homogenizer (IKA Werke GmbH and Co. KG, Staufen, Germany) and subsequently treated as per plasma samples.


Samples were analyzed on an Agilent 1100 series HPLC system (Agilent Technologies, Santa Clara, Calif., USA), configured as described above, using the method of Leyton J, Smith G, Zhao Y, et al. [18F]fluoromethyl-[1, 2-2H4]-choline: a novel radiotracer for imaging choline metabolism in tumors by positron emission tomography. Cancer Res. 2009; 69:7721-7728. A μBondapak C18 HPLC column (Waters, Milford, Mass., USA; 7.8×3000 mm), stationary phase and a mobile phase comprising of Solvent A (vide supra) and Solvent B (acetonitrile/water/ethanol/acetic acid/1.0 M ammonium acetate/0.1 M sodium phosphate (400/400/68/44/88/10)), delivered at a flow rate of 3 mL/min were used for analyte separation. The gradient was set as follows: 0% B for 5 min; 0% to 100% B in 10 min; 100% B for 0.5 min; 100% to 0% B in 2 min; 0% B for 2.5 min.


PET Imaging Studies

Dynamic 11C-choline, 11C-D4-choline and 18F-D4-choline imaging scans were carried out on a dedicated small animal PET scanner (Siemens Inveon PET module, Siemens Medical Solutions USA, Inc., Malvem, Pa., USA) following a bolus i.v. injection in tumor-bearing mice of either ˜3.7 MBq for 18F studies, or ˜18.5 MBq for 11C. Dynamic scans were acquired in list mode format over 60 min. The acquired data were then sorted into 0.5 mm sinogram bins and 19 time frames for image reconstruction (4×15 s, 4×60 s, and 11×300 s), which was done by filtered back projection. For input function analysis, data were sorted into 25 time frames for image reconstruction (8×5 s, 1×20 s, 4×40 s, 1×80 s, and 11×300 s). The Siemens Inveon Research Workplace software was used for visualization of radiotracer uptake in the tumor; 30 to 60 min cumulative images of the dynamic data were employed to define 3-dimensional (3D) regions of interest (ROIs). Arterial input function was estimated as follows: a single voxel 3D ROI was manually drawn in the center of the heart cavity using 2 to 5 min cumulative images. Care was taken to minimize ROI overlap with the myocardium. The count densities were averaged for all ROIs at each time point to obtain a time versus radioactivity curve (TAC). Tumor TACs were normalized to injected dose, measured by a VDC-304 dose calibrator (Veenstra Instruments, Joure, The Netherlands), and expressed as percentage injected dose per mL tissue. The area under the TAC, calculated as the integral of % ID/mL from 0-60 min, and the normalized uptake of radiotracer at 60 min (% ID/mL60) were also used for comparisons.


Biodistribution Studies


11C-choline, 11C-D4-choline (˜18.5 MBq) and 18F-D4-choline (˜3.7 MBq) were each injected via the tail vein of anaesthetized BALB/c nude mice. The mice were maintained under anesthesia and sacrificed by exsanguination via cardiac puncture at 2, 15, 30 or 60 min post radiotracer injection to obtain blood, plasma, heart, lung, liver, kidney and muscle. Tissue radioactivity was determined on a gamma counter (Cobra II Auto-Gamma counter, Packard Biosciences Co, Pangbourne, UK) and decay corrected. Data were expressed as percent injected dose per gram of tissue.


Statistics

Data were expressed as mean±standard error of the mean (SEM), unless otherwise shown. The significance of comparison between two data sets was determined using Student's t test. ANOVA was used for multi-parametric analysis (Prism v5.0 software for windows, GraphPad Software, San Diego, Calif., USA). Differences between groups were considered significant if P≦0.05.


Results
Deuteration Leads to Enhanced Renal Radiotracer Uptake

Time course biodistribution was performed in non-tumor-bearing male nude mice with 11C-choline, 11C-D4-choline and 18F-D4-choline tracers. FIG. 20 shows tissue distribution at 2, 15, 30 and 60 min. There were minimal differences in tissue uptake between the three tracers over 60 min, with uptake values in broad agreement with data previously published for 18F-choline and 18F-D4-choline (DeGrado T R, Baldwin S W, Wang S, et al. Synthesis and evaluation of (18)F-labeled choline analogs as oncologic PET tracers. J Nucl Med. 2001; 42:s1805-1814; Smith G, Zhao Y, Leyton J, et al. Radiosynthesis and pre-clinical evaluation of [(18)F]fluoro-[1,2-(2)H(4)]choline. Nucl Med Biol. 2011; 38:39-51). In all tracers there was rapid extraction from blood, with the majority of radioactivity retained within the kidneys, evident as early as 2 min post injection. Deuteration of 11C-choline led to a significant 1.8-fold increase in kidney retention over 60 min (P<0.05; FIG. 20A), with a 3.3-fold increase in kidney retention observed for 18F-D4-choline when compared to 11C-choline at this time point (P<0.01). There was a trend towards increased urinary excretion for 11C-D4-choline and 18F-D4-choline, in comparison to the nature identical tracer, 11C-choline, although this increase did not reach statistical significance.


Deuteration of 11C-choline Results in Modest Resistance to Oxidation In Vivo

Tracer metabolism in tissues and plasma was performed by radio-HPLC (FIG. 21). Peaks were assigned as choline, betaine, betaine aldehyde and phosphocholine, using enzymatic (alkaline phosphatase and choline oxidase) methods to determine their identity (FIGS. 27 and 28, respectively) (Leyton J, Smith G, Zhao Y, et al. [18F]fluoromethyl-[1, 2-2H4]-choline: a novel radiotracer for imaging choline metabolism in tumors by positron emission tomography. Cancer Res. 2009; 69:7721-7728).


In the liver, both 11C-choline and 11C-D4-choline were rapidly oxidized to betaine (FIG. 21A), with 49.2±7.7% of 11C-choline radioactivity already oxidized to betaine by 2 min. Deuteration of 11C-choline provided significant protection against oxidation in the liver at 2 min post injection, with 24.5±2.1% radioactivity as betaine (51.2% decrease in betaine levels; P=0.037), although this protection was lost by 15 min. Notably, a high proportion of liver radioactivity (˜80%) was present as phosphocholine by 15 min with 18F-D4-choline. This corresponded to a much reduced liver-specific oxidation when compared to the two carbon-11 tracers (15.0±3.6% of radioactivity as betaine at 60 min; P=0.002).


In contrast to the liver, deuteration of 11C-choline resulted in protection against oxidation in the kidney over the entirety of the 60 min time course (FIG. 21B). With 11C-D4-choline there was a 20-40% decrease in betaine levels over 60 min when compared to 11C-choline (P<0.05), corresponding to a proportional increase in phosphocholine (P<0.05). 18F-D4-choline was more resistant to oxidation in the kidney than both carbon-11 labeled choline tracers. There was a relationship between levels of radiolabeled phosphocholine and kidney retention when data from all three tracers were compared (R2=0.504; FIG. 29). In the plasma, the temporal levels of betaine for both 11C-choline and 11C-D4-choline were almost identical; it should be noted that total radioactivity levels were low for all radiotracers. At 2 min, 12.1±2.6% and 8.8±3.8% of radioactivity was in the form of betaine for 11C-choline and 11C-D4-choline respectively, rising to 78.6±4.4% and 79.5±2.9% at 15 min. Betaine levels were significantly reduced with 18F-D4-choline, with 43.7±12.4% of activity present as betaine at 15 min. A further increase in plasma betaine was not observed with 18F-D4-choline over the remainder of the time course.


Fluorination Protects Against Choline Oxidation in Tumor


11C-choline, 11C-D4-choline and 18F-D4-choline metabolism were measured in HCT116 tumors (FIG. 22). With all tracers, choline oxidation was greatly reduced in the tumor in comparison to levels in the kidney and liver. At 15 min, both 11C-D4-choline and 18F-D4-choline had significantly more radioactivity corresponding to phosphocholine than 11C-choline; 43.8±1.5% and 45.1±3.2% for 11C-D4-choline and 18F-D4-choline respectively, in comparison to 30.5±4.0% for 11C-choline (P=0.035 and P=0.046 respectively). By 60 min, the majority of radioactivity was phosphocholine for all three tracers, with phosphocholine levels increasing in the order of 11C-choline<11C-D4-choline<18F-D4-choline. There was no difference in the tumor metabolic profile for 11C-choline and 11C-D4-choline at 60 min, although reduced choline oxidation was observed for 18F-D4-choline; 14.0±3.0% betaine radioactivity with 18F-D4-choline compared with 28.1±2.9% for 11C-choline (P=0.026).


Choline Tracers have Similar Sensitivity for Imaging Tumors by PET


Despite the high systemic stability of 18F-D4-choline, tumor radiotracer uptake in mice by PET was no higher than with 11C-choline or 11C-D4-choline (FIG. 23). FIG. 23A shows typical (0.5 mm) transverse PET image slices showing accumulation of all three tracers in HCT116 tumors. For all three tracers there was heterogeneous tumor uptake, but tumor signal-to-background levels were identical; confirmed by normalized uptake values at 60 min and values for the tumor area under the time verses radioactivity curve (data not shown). It should be noted that the PET data represent total radioactivity. In the case of 11C-choline or 11C-D4-choline, a significant proportion of this radioactivity is betaine (FIG. 22).


Tumor Tracer Kinetics

Despite there being no difference in overall tracer retention in the tumor, the kinetic profiles of tracer uptake varied between the three choline tracers, detected by PET (FIG. 23B). The kinetics for the three tracers were characterized by rapid tumor influx over the initial 5 min, followed by stabilization of tumor retention. Initial delivery of 18F-D4-choline over the first 14 min of imaging was higher than for both 11C-choline and 11C-D4-choline (expanded TAC for initial 14 min shown in FIG. 30). Slow wash-out of activity was observed with both 18F-D4-choline and 11C-D4-choline between 30 and 60 min, in contrast to the gradual accumulation detected with 11C-choline. Parameters for the irreversible trapping of radioactivity in the tumor, Ki and k3, were calculated from a two-tissue irreversible model, using metabolite-corrected TAC from the heart cavity as input function (FIGS. 24A and B). A double input (DI) model, accounting for the contribution of metabolites to the tissue TAC, was used for kinetic analysis, described in supplemental data. There was no significant difference in flux constant measurements between deuterated and undeuterated 11C-choline. Addition of methylfluoride, however, resulted in 49.2% (n=3; P=0.022) and 75.2% (n=3; P=0.005 decreases in Ki and k3, respectively; i.e., when 18F-D4-choline was compared to 11C-D4-choline. Ki′ values were similar between all three tracers: 0.106±0.026; 0.114±0.019; 0.142±0.027 for 11C-choline, 11C-D4-choline and 18F-D4-choline respectively. It is possible that intracellular betaine formation (not just presence of betaine in the extracellular space) led to a higher than expected irreversible uptake; there was a significant 388 and 230% increase in the ratio of betaine:phophocholine at 15 and 60 min respectively (P=0.045 and 0.036) with 11C-choline in comparison to 18F-D4-choline (FIG. 5C).



18F-D4-choline Shows Good Sensitivity for the PET Imaging of Prostate Adenocarcinoma and Malignant Melanoma


Having confirmed that 18F-D4-choline is a more stable choline analogue for in vivo studies, with good sensitivity for the imaging of colon adenocarcinoma, it was desired to evaluate its suitability for cancer detection in other models of human cancer including malignant melanoma A375 and prostate adenocarcinoma PC3-M. In vitro uptake of 18F-D4-choline was similar in the three cell lines over 30 min (FIG. 31), relating to near-identical levels of choline kinase expression (FIG. 31 insert). Retention of radioactivity was shown to be choline kinase-dependent as treatment of cells with the choline transport and choline kinase inhibitor, hemicholinium-3, resulted in >90% decrease in intracellular tracer radioactivity in all three cell lines. Similar intracellular trapping of 18F-D4-choline in these cancer models were translated to their uptake in vivo (FIG. 25A)), showing similar values for flux constant measurements and PET imaging variables (Supplemental Table 1). There was a trend towards increased tumor retention of 18F-D4-choline in the order of A375<HCT116<PC3-M; reflected by the expression of choline kinase in these lines (FIG. 25C). There was no discernable difference in tumor metabolite profiles between the three cell cancer models at either 15 or 60 min post injection (FIG. 25B).


Tumor Size Affects 18F-D4-choline Uptake and Retention but not Tumor Pharmacokinetics


For PET imaging, tumors were grown to 100 mm3 prior to imaging. One small cohort of animals with implanted PC3-M xenografts were, however, imaged when the tumor size had reached 200 mm3 (See FIG. 32 for typical transverse PET images). These tumors showed a distinct pattern of 18F-D4-choline uptake around the tumor rim, corresponding to a substantial decrease in tumor radioactivity when compared to smaller PC3-M tumors (FIG. 26). As with HCT116 tumors, maximal tumor-specific radioactivity was achieved within 5 min of tracer injection in both PC3-M cohorts, followed by a plateau. The magnitude of radiotracer retention at 60 min was substantially higher in the smaller tumors, with a normalized uptake value of 1.97±0.07% ID/mL versus 0.82±0.12% ID/mL in the larger tumors (2.4-fold increase; P=0.0002; n=3-5). Analysis of tumor uptake, taking the maximal voxel radioactivity value from the tumor ROI, resulted in a smaller difference in tracer uptake at 60 min, with an % ID/mLmax of 4.75±0.38 measured in the ˜100 mm3 tumor in comparison to 3.34±0.08% ID/mLmax measured in the ˜200 mm3 tumor (1.4-fold increase; P=0.019; n=3-5). Interestingly, there was no significant change in the kinetic parameters measuring the irreversible trapping of radioactivity, Ki and k3, between both tumor cohorts.


Kidney retention increased in the order of 11C-choline<11C-D4-choline<18F-D4-choline over the 60 min time course (FIG. 20), with total kidney radioactivity shown to be proportional to the % radioactivity retained as phosphocholine (FIG. 29; R2=0.504). Protection against choline oxidation by deuteration of 11C-choline was shown to be tissue specific, with a decrease in betaine radioactivity measured in the liver at just 2 min post injection when compared to 11C-choline (FIG. 21).


Despite systemic protection against choline oxidation with 18F-D4-choline, the reduction in the rate of choline oxidation was much more subtle in implanted HCT116 tumors (FIG. 22). At 15 min post injection there were 43.6% and 47.9% higher levels of radiolabeled-phosphocholine when 11C-D4-choline and 18F-D4-choline, respectively, were injected relative to 11C-choline. By 60 min there was no difference in phosphocholine levels between the three tracers, although there was a significant decrease in betaine-specific radioactivity with 18F-D4-choline. This equilibration of phosphocholine-specific activity can be explained by a saturation effect, with parent tracer levels reduced to a minimum by 60 min, severely limiting substrate levels available for choline kinase activity. Lower betaine levels were observed in the tumor with all three tracers over the entire time course when compared to liver and kidney, likely resulting from a lower capacity for choline oxidation or increased washout of betaine.


Comparison of the three choline radiotracers by PET showed no significant differences in overall tumor radiotracer uptake and hence sensitivity (FIG. 23) despite large changes observed in other organs. Initial tumor kinetics (at time points when metabolism was lower), however, varied between tracers, with 18F-D4-choline characterized by rapid delivery over ˜5 min, followed by slow wash-out of activity from the tumor. This compared to the slower uptake, but continuous tumor retention of 11C-choline. At 60 min a 2.7-fold and 4.0-fold higher un-metabolized parent tracer was seen with 18F-D4-choline in tumor compared to 11C-choline and 11C-D4-choline, respectively, (FIG. 22). Deuteration did not, however, alter total tumor radioactivity levels and the modeling approach used did not distinguish between different intracellular species. While all tracers were converted intracellularly to phosphocholine, the higher rate constants for intracellular retention (Ki and k3; FIGS. 24A and B) of 11C-choline and 11C-D4-choline, compared to 18F-D4-choline were explained by the rapid conversion of the non-fluorinated tracers to betaine within HCT116 tumors, indicating greater specificity with 18F-D4-choline. Compared to 18F-D4-choline, the tumor betaine-to-phosphocholine metabolite ratio increased by 388% (P=0.045) and 259% (P=0.061, non-significant) for 11C-choline and 11C-D4-choline, respectively (FIG. 24C).


Example 22
General

Materials were used as purchased without further purification. 1,2-2H4-Dimethylethanolamine (DMEA) was a custom synthesis by Target Molecules Ltd (Southampton, UK). Water for irrigation was from Baxter (Deerfield, Ill., USA) and soda lime was purchased from VWR (Lutterworth, Leicestershire, UK). 0.9% sodium chloride for injection was from Hameln pharmaceuticals Ltd (Gloucester, UK) a 0.045% solution of NaCl was prepared from this stock and water for irrigation. Lithium aluminium hydride (0.1 M in THF) and hydriodic acid (57%) were from ABX (Radeburg, Germany). Methylene ditosylate was obtained from the Huayi Isotope Company (Toronto, Canada). All other chemicals were from Sigma-Aldrich Co. Ltd (Poole, Dorset, UK). For 11C-methylations on the iPhase 11C-PRO, iPhase disposable synthesis kits were obtained from iPhase Technologies Pty Ltd (Melbourne, Australia). For 18F-fluoromethylations on the GE FASTlab (GE Healthcare, Chalfont St. Giles, UK) the partly assembled GE FASTlab cassette contained a FASTlab water bag, N2 filter, pre-conditioned QMA cartridge and reaction vessel. Waters Sep-Pak Accell CM light, tC18 light and tC18 Plus cartridges were obtained from Waters Corporation (Milford, Ma., USA).


Synthesis of 11C-Choline and 11C-[1,2-2H4]-choline



11C-Methyl iodide was prepared using a standard wet chemistry method. Briefly, 11C-carbon dioxide was transferred to the iPhase platform via a custom attached cryogenic trap and reduced to 11C-methane using lithium aluminium hydride (0.1 M in THF) (200 uL) over 1 min at RT. Concentrated hydroiodic acid (200 μL) was then added to the reactor vessel and the mixture heated to 140° C. for 1 min. 11C-methyl iodide was then distilled through a short column containing soda lime and phosphorus pentoxide desiccant into a 2 mL stainless steel loop containing the precursor dimethylethanolamine or 1,2-2H4-dimethylethanolamine (201). The methylation reaction was allowed to proceed at room temperature for 2.5 min. The crude product was then flushed on to a CM cartridge using ethanol (20 mL) at a flow rate of 5 mL/min. The CM cartridge had previously been pre-conditioned with 0.045% sodium chloride (5 mL) then water (5 mL). The CM cartridge was then washed with aqueous ammonia (0.08%, 15 mL) then water (10 mL). The choline product was then eluted from the cartridge using sodium chloride solution (0.045%, 10 mL).


Synthesis of 18F-fluoromethyl-[1,2-2H4]-choline


The system was configured with an eluent vial comprising of 1:4 K2CO3 solution in water:Kryptofix K222 solution in acetonitrile (1.0 mL), 180 mg K2CO3 in water (10.0 mL) and 120 mg Kryptofix K222 in acetonitrile (10.0 mL), methylene ditosylate (4.2-4.4 mg) in acetonitrile (2% water; 1.25 mL), precursor 1,2-2H4-dimethylethanolamine (150 μl) in anhydrous acetonitrile (1.4 mL).


Fluorine-18 drawn onto system and immobilised on Waters QMA light cartridge then eluted with 1 mL of a mixture of carbonate and kryptofix into the reaction vessel. After the K[18F]F/K222/K2CO3 drying cycle was complete, methylene ditosylate in acetonitrile (2% water; 1.25 mL) was added and reaction vessel heated to 110° C. for minutes. The reaction was quenched with water (3 mL) and the resulting mixture was passed through both t-C18 light and t-C18 plus cartridges (pre-conditioned with acetonitrile and water; 2 mL each); 15% acetonitrile in water was then passed through the cartridges. After completion of the clean-up cycle, methylene ditosylate was trapped on the t-C18 light cartridge and 18F-fluoromethyl tosylate (together with 18F-tosyl fluoride) was retained on the t-C18 plus, with other reactants going to waste. The washing cycles ethanol→vacuum→nitrogen were employed to clean the reaction vessel after this first stage of radiosynthesis. The reaction vessel and the t-C18 plus cartridge with immobilized 18F-fluoromethyl tosylate were then simultaneously dried under a stream of nitrogen. 18F-fluoromethyl tosylate was then eluted from the t-C18 plus cartridge with 150 μl of 1,2-2H4-dimethylethanolamine in 1.4 mL of acetonitrileinto the reaction vessel. The reactor vessel was then heated to 110° C. for 15 minutes then cooled and the reaction vessel contents washed with water on to a CM cartridge (conditioned with 2 mL water). The cartridge was washed by withdrawing ethanol from the bulk ethanol vial and passing it through CM cartridge; the washing cycle was repeated once followed by 0.08% ammonia solution (4.5 mL). The CM cartridge then was subjected to final washes with ethanol followed by water. The product, 18F-fluoro-[1,2-2H2]choline, was washed off the CM cartridge with 0.09% sodium chloride solution (4.5 mL) to afford 18F-fluoro-[1,2-2H2]choline in sodium chloride buffer as the final formulated product.


Assessment of Chemical/Radiochemical Purity


11C-Choline, 11C-[1,2-2H4]-choline and 18F-fluoro-[1,2-2H2]choline were analyzed for chemical/radiochemical purity on a Metrohm ion chromatography system (Runcorn, UK) with a Metrosep C4 cation column (250×4.0 mm) attached. The mobile phase was 3 mM Nitric acid:Acetonitrile (75:25 v/v) running in isocratic mode at 1.5 mL/min. All radiotracers were >95% radiochemical purity after formulation.


Kinetic Analysis in HCT116 Tumors

A 2-tissue irreversible compartmental model was employed to fit the TACs, as has been previously established for 11C-choline (Kenny L M, Contractor K B, Hinz R, et al. Reproducibility of [11C]choline-positron emission tomography and effect of trastuzumab. Clin Cancer Res. Aug. 15 2010; 16(16):4236-4245; and Sutinen E, Nurmi M, Roivainen A, et al. Kinetics of [(11)C]choline uptake in prostate cancer: a PET study. Eur J Nucl Med Mol. Imaging. March 2004; 31(3):317-324). An estimate of the whole blood TAC (wbTAC(t)) was derived from the PET image itself, as described above. As the wbTAC was obtained from one voxel only it was relatively noisy. Therefore it was fitted with a sum of 3 exponentials from the peak on and the fitted function was used as input function in the kinetic modeling (after metabolite correction, see below). The parent fraction values, pf, were calculated from plasma metabolite analysis: at 2, 15, 30 and 60 minutes they were [0.96, 0.55, 0.47, 0.26] for 18F-D4-choline, [0.92, 0.25, 0.20, 0.12] for 11C-choline and [0.91, 0.18, 0.08, 0.03] for 11C-D4-choline, respectively. The pf values were fitted to a sum of two exponentials with the constraint pf(t=0)=1 to obtain the function pf(t). The parent whole blood TAC wbTACPAR(t) was then computed by multiplying wbTAC(t) and pf(t) and used as input function to estimate the parameters K1 (mL/cm3/min), k2 (1/min), k3 (1/min) and Vb (unitless). The steady state net irreversible uptake rate constant Ki (mL/cm3/min) was calculated from the estimated microparameters as K1k3/(k2+k3). Because the quality of fits obtained using the wbTACPAR(t) as only input function to the model was poor, and because 18F-D4-choline, 11C-choline and 11C-D4-choline are quickly metabolized in vivo in the mouse, a double input (DI) model accounting for the contribution of metabolites to the tissue TAC was also considered (Huang S C, Yu D C, Barrio J R, et al. Kinetics and modeling of L-6-[18F]fluoro-dopa in human positron emission tomographic studies. J Cereb Blood Flow Metab. November 1991; 11(6):898-913). In the DI model the metabolite whole blood TAC wbTACMET(t) computed as wbTAC(t)x[1-pf(t)] together with wbTACPAR(t) was employed as input function; the parent tracer was modeled with a 2-tissue irreversible model whereas a simple 1-tissue reversible model was used to describe the metabolite kinetics, thus computing the metabolite influx and efflux K1′ and k2′ in addition to the parameters estimated for the parent. The standard Weighted Non-Linear Least Squares (WNLLS) was used as estimation procedure. WNLLS minimizes the Weighted Residual Sum of Squares (WRSS) function










WRSS


(
p
)


=




i
=
1

n





w
i



[



C


(


t
i

,
p

)


MODEL

-

C


(

t
i

)



]


2






(
A
)







with C(ti) and ti indicating respectively the decay-corrected concentration computed from the PET image and the mid-time of the i-th frame and n denoting number of frames. In Eq.1 weights wi were set to










Δ
i



C


(

t
i

)




exp


(

λ






t
i


)







(
B
)







with Δi and 2 representing the duration of the i-th frame and the half-life of 18F (for 18F-D4-choline) or 11C (for 11C-choline and 11C-D4-choline) (Tomasi G, Bertoldo A, Bishu S, Unterman A, Smith C B, Schmidt K C. Voxel-based estimation of kinetic model parameters of the L-[1-(11)C]leucine PET method for determination of regional rates of cerebral protein synthesis: validation and comparison with region-of-interest-based methods. J Cereb Blood Flow Metab. July 2009; 29(7):1317-1331). WNLLS estimation was performed with the Matlab function lsqnonlin; parameters were constrained to be positive but no upper bound was applied.


Supplemental Table 1.


Kinetic parameters from dynamic 18F-D4-choline PET in tumors. Decay-corrected uptake values at 60 min (NUV60) and the area under the curve (AUC) were taken from tumor TACs. Flux constant measurements, K1′, Ki and k3 were obtained by fitting tumor TAC and derived input function, corrected for radioactive plasma metabolites of 18F-D4-choline, to a 2-tissue irreversible model of tracer delivery and retention. Mean values (n=3)±SEM are shown.



















NUV60
AUC
K1
Ki
k3





















HCT116
1.81 ± 0.11
114.5 ± 7.0
0.142 ± 0.027
0.008 ± 0.001
0.039 ± 0.003


A375
1.71 ± 0.14
107.3 ± 7.7
0.111 ± 0.021
0.006 ± 0.002
0.030 ± 0.008


PC3-M
1.97 ± 0.07
121.3 ± 3.1
0.090 ± 0.007
0.009 ± 0.002
0.040 ± 0.006










All patents, journal articles, publications and other documents discussed and/or cited above are hereby incorporated by reference.

Claims
  • 1. A compound of Formula (III):
  • 2. The compound according to Claim 1 wherein C* is 11C, 13C, or 14C.
  • 3. The compound according to Claim 1 wherein C* is 11C; X and Y are each hydrogen; and Z is F.
  • 4. The compound according to Claim 1 wherein C* is 11C; X, Y and Z are each hydrogen H; R1, R2, R3, and R4 are each deuterium (D); and R5, R6, and R7 are each hydrogen. 5. A pharmaceutical composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier or excipient.
  • 6. A pharmaceutical composition comprising a compound of claim 2 and a pharmaceutically acceptable carrier or excipient.
  • 7. A pharmaceutical composition comprising a compound of claim 3 and a pharmaceutically acceptable carrier or excipient.
  • 8. A pharmaceutical composition comprising a compound of claim 4 and a pharmaceutically acceptable carrier or excipient.
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2011/052275 9/20/2011 WO 00 3/18/2013
Provisional Applications (2)
Number Date Country
61384895 Sep 2010 US
61531119 Sep 2011 US