AZ13198083, a Novel Histamine Type-3 Receptor R - ACS Publications

Jan 23, 2018 - [Carbonyl-11C]6 displayed the most favorable in vivo kinetics and brain uptake, with an early peak in the striatal time−activity curv...
0 downloads 8 Views 778KB Size
Subscriber access provided by READING UNIV

Letter

Development of [carbonyl-11C]AZ13198083, a novel histamine type-3 receptor radioligand with favorable kinetics Kenneth Dahl, Ryuji Nakao, Nahid Amini, Mohammad Mahdi Moein, Sjoerd J Finnema, Jonas Malmquist, Katarina Varnas, and Magnus Schou ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.7b00493 • Publication Date (Web): 23 Jan 2018 Downloaded from http://pubs.acs.org on January 24, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Chemical Neuroscience is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 7 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

Development of [carbonyl-11C]AZ13198083, a novel histamine typetype-3 receptor radioligand with favorable kinetics Kenneth Dahl,† Ryuji Nakao,† Nahid Amini,† Mohammad Mahdi Moein,† Sjoerd Finnema,† Jonas Malmquist‡, Katarina Varnäs,† and Magnus Schou*, †, ‡ †Department

of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden ‡PET Science Centre, Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Karolinska Institutet, S-171 76 Stockholm, Sweden. KEYWORDS: PET imaging, radioligand, receptor, histamine, carbon-11, carbonylation

ABSTRACT: The histamine subtype-3 receptor (H3R) is implicated in a range of central nervous system disorders and several radioligands have been developed for H3R positron emission tomography imaging. However, a limitation of currently used PET radioligands for H3R is the slow binding kinetics in high density brain regions. To address this, we herein report the development of three novel candidate H3 R radioligands, namely, [carbonyl-11C]AZ13153556 ([carbonyl-11C]4), [carbonyl11 11 11 11 C]AZD5213([carbonyl- C]5), and [carbonyl- C]AZ13198083 ([carbonyl- C]6), and their subsequent preclinical evaluation in non-human primates (NHP). Radioligands [carbonyl-11C]4-6 were produced and isolated in high radioactivity (>1000 MBq), radiochemical purity (> 99%), and moderate molar activity (19-28 GBq/µmol at time of injection) using a palladium-mediated 11Caminocarbonylation protocol. All three radioligands showed high brain permeability as well as a regional brain radioactivity distribution in accordance with H3R expression (striatum > cortex > cerebellum). [Carbonyl-11C]6 displayed the most favorable in vivo kinetics and brain uptake, with an early peak in the striatal time-activity curve followed by a progressive washout from the brain. The specificity and on-target kinetics of [carbonyl-11C]6 were next investigated in pretreatment and displacement studies. After pretreatment or displacement with 5 (0.1 mg/kg), a uniformly low distribution of radioactivity across the NHP brain was observed. Collectively, this work demonstrates that [carbonyl-11C]6 is a promising candidate for H3R imaging in human subjects.

The histamine type-3 receptor (H3R) is widely expressed in the central nervous system (CNS) where it plays an important role in regulating the release of the neurotransmitters histamine, dopamine and noradrenaline. The highest H3R densities in brain are found in regions involved in cognitive processes (e.g. cerebral cortex and basal ganglia). H3Rs have been implicated in a range of disorders including Alzheimer’s disease, schizophrenia, obesity and diabetes.1-5 Positron emission tomography (PET) is a sensitive noninvasive imaging technique. The PET technology relies upon the intravenous administration of a radiolabeled imaging agent (radioligand) that serves as a molecular probe to investigate biological processes in vivo.6-9 A number of H3R PET radioligands have been reported,10-20 but only a few have shown promise in vivo. Of these, three have so far been translated for human use, namely, [11C]MK8278 ([11C]1)16, [11C]GSK189254 ([11C]2)13, 19, and [11C]TASP457 ([11C]3)20 (Figure 1, top). A common feature with these radioligands is the slow binding kinetics in high density brain regions, which in turn, leads to high variance in estimates of binding parameters. Figure 1. Top: PET radioligands translated for in human imaging of the H3R. Bottom: Structures of our previous and current efforts to H3R radioligand development.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 7

Table 1. Radiosynthesis of three novel histamine subtype-3 receptor PET radioligands ([carbonyl-11C]4-6) using Pd-mediated 11Ccarbonylative coupling of a bromoarene with primary amines.[a]

Radioligand ([carbonyl-11C]4-6) [carbonyl-11C]4 [carbonyl-11C]5 [carbonyl-11C]6

Amine (8a-c)

8a, NH2CH3 8b, NH3 8c, NH2C2H5

Radiochemical yield (%)b 80 96 95

Radiochemical purity (%)c >99 >99 >99

Synthesis (min)d

time

41 32 38

Molar Activity at TOI (GBq/µmol)e 19 28 23

[a] Conditions: Bromo-precursor (7, 20 µmol), amine (8a-c, 200 µmol), Pd2[cinnamyl-π]Cl2 (14 µmol), xantphos (14 µmol), anhydrous THF (Tetrahydrofuran, 500 µL), 100ᵒC, 5 min. [b] Decay-corrected and non-isolated radiochemical yield relative to the amount of [11C]CO delivered to the reaction vessel. [c] Radiochemical purity of isolated products measured by analytical highperformance liquid chromatography. [d] The overall synthesis time, including 11C-labeling, purification, formulation and sterile filtration. [e] Molar Activity of isolated products measured by analytical high-performance liquid chromatography and calculated at TOI (time-of-injection).

Figure 2. Time-activity curves for the concentration of radioactivity in various monkey brain regions as a function of time following intravenous injection of [carbonyl-11C]4 (left), [carbonyl-11C]5 (middle), [carbonyl-11C]6 (right) at baseline. STR, striatum; PFC, prefrontal cortex; OC, occipital cortex; CER, cerebellum.

We previously reported the development of [methyl11 C]AZ13153556 ([methyl-11C]4),21 a H3R selective PET radioligand that showed good imaging properties in vivo, including high brain uptake, reversible binding and negligible nonspecific binding in non-human primate (NHP) brain. However, in common with [11C]1-3, [methyl-11C]4 also displayed relatively slow binding kinetics in regions with high H3R density. Herein, we report the radiosyntheses and preclinical evaluation of two novel H3R PET radioligands, [carbonyl11 C]AZD5213 ([carbonyl-11C]5) and [carbonyl11 C]AZ13198083 ([carbonyl-11C]6), as well as, [carbonyl11 C]4 (Figure 1, bottom), in order to explore the impact of amide substituents on brain kinetics in vivo.

RESULTS AND DISCUSSION Radiochemistry. 11C-labeled carbon monoxide ([11C]CO) has been increasingly recognized as an important 11C-labeling agent for radiopharmaceutical research and production.22-26 No-carrier-added (nca) [11C]CO is commonly obtained from cyclotron produced [11C]carbon dioxide ([11C]CO2) by metalmediated on-line reduction27-29, and can be employed in many metal-mediated carbonylation reactions, giving rise to a wide library of radiolabeled compounds. Various ketones, amides, esters, carboxylic acids, ureas and carbamates are examples of functional groups accessible via metal-mediated 11Ccarbonylation reactions.22-24 As neither 5 or 6 have a terminal methyl group present, the standard radiomethylation protocol

ACS Paragon Plus Environment

Page 3 of 7 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience was abandoned for a high throughput labeling strategy based on [11C]CO (Table 1). Radioligands [carbonyl-11C]4-6 were efficiently prepared by [11C]aminocarbonylation of the corresponding aryl bromide precursor.30-31 Reaction conditions and production data are shown in Table 1. The reaction was conducted using 7 as substrate, [11C]CO as the 11C-source, an amine (8a-c), and Pd2(π-cinnamyl)Cl2-xantphos as catalyst with anhydrous tetrahydrofuran (THF) as solvent. In this study, nca [11C]CO (radiochemical yield (RCY) ~ 70%) was produced by on-line reduction of in-target produced [11C]CO2 over heated (850ᵒC) molybdenum.27 The [11C]CO was next reacted (100ᵒC for 5 min) with the precursor-solution in a steel reaction chamber (µ-autoclave, V = 250 µL) submerged in an oil bath.32 When the 11C-carbonylative step was completed, the mixture was automatically eluted into a receiving vial for further purification. Radioligands [carbonyl-11C]4-6 were produced in radioactivities >1 GBq (27 mCi, isolated, formulated and sterile filtered) and >99% radiochemical purity (RCP) using the present conditions. The overall synthesis time was 32-41 min and the molar activity (Am) 19-28 GBq/µmol (513-757 Ci/mmol) at the time-of-injection. The obtained Am during this study was 10-fold lower compared to our previous study with [methyl-11C]4.21 However, it is on a similar level as other radioligands prepared from [11C]CO in our laboratory.29 Preclinical PET imaging. With the radioligands in hand, we conducted dynamic PET imaging studies in female cynomolgus monkeys. The aim of the initial NHP studies was to evaluate the suitability of [carbonyl-11C]4-6 as PET radioligands with regards to; (i) blood brain barrier (BBB) permeability, (ii) regional brain distribution, and (iii) brain kinetics. To facilitate a direct comparison, each radioligand ([carbonyl11 C]4-6) was injected intravenously in the same monkey on the same experimental day. Following a 123 min emission measurement, PET images were reconstructed and region of interests (ROIs) were delineated based on magnetic resonance (MR) images. Time-radioactivity curves (TACs) for the regional brain distribution of [carbonyl-11C]4-6 were next generated (Figure 2). All three radioligands rapidly crossed the BBB and showed a regional brain distribution consistent with H3R expression.33-34 The regional uptake was most prominent in H3R-rich regions, such as striatum, while moderate in the cortex and significantly lower in cerebellum (Figure 2). Importantly, we observed a significant difference in both brain uptake and kinetics between the three radioligands. For example, a peak in the striatal TAC was not observed during the entire PET measurement with [carbonyl-11C]5 (Figure 2, middle), whereas TAC peaks were obtained early with both [carbonyl-11C]4 and [carbonyl-11C]6. The subsequent washout from striatum was on the other hand considerably more rapid with [carbonyl-11C]6 than with [carbonyl-11C]4 (Figure 2). The faster brain kinetics observed with [carbonyl-11C]6 could, at least in part, be explained by its 4-fold lower affinity for the H3R in vitro (4, IC50 = 6.4 nM; 6, IC50 = 24 nM). During the PET measurements, radiolabeled metabolites in plasma were analyzed using radio-HPLC (see supporting information for further information). Interestingly, [carbonyl-11C]6 was more rapidly metabolized compared to [carbonyl-11C]4. This likely also contributes to the accelerated wash-out of [carbonyl-11C]6 from brain by shifting the equilibrium between radioligand concentration in brain and plasma towards plasma. It is noteworthy that [carbonyl-11C]4 displayed a slightly better

in vivo binding kinetics in comparison to our previously reported 11C-methylated analogue, [methyl-11C]4.21 One possible explanation for this may be the lower molar activity obtained in the current study, (i.e. a mass effect). Such mass effects are not uncommon for high affinity PET radioligands and was indeed reported for H3R imaging in human subjects with [11C]GSK189254, where a dose lower than 0.003 µg/kg of body weight was recommended to achieve less than 5% occupancy induced by the carrier.19 Based on its favorable kinetics and regional brain distribution, [carbonyl-11C]6 was chosen for further evaluation. First a pretreatment study was performed, in which the selective H3R antagonist 5 was infused at a dose (0.1 mg/kg) known to occupy >95% of H3R in human brain.35 TACs for the regional brain distribution of [carbonyl-11C]6 were generated at baseline (Figure 3, top) and at pretreatment conditions (Figure 3, bottom). Gratifyingly, pre-treatment with 5 almost completely inhibited the specific binding of [carbonyl-11C]6 in the examined H3R rich regions, leading to a uniformly low distribution of radioactivity in the NHP brain. The strong effect of 5 on the regional distribution of [carbonyl-11C]6 supports the view that binding of this radioligand is predominantly specific to the H3R. Summed PET-MR images (3-123 min) after intravenous administration of [carbonyl-11C]6 in monkey brain at baseline (Figure 4, top) and pre-treatment conditions (Figure 4, bottom) are shown in Figure 4. Finally, the binding of [carbonyl-11C]6 was characterized by a displacement study in which 5 was administered 60 minutes after [carbonyl-11C]6. TACs of [carbonyl-11C]6 were generated at baseline (Figure 5, top) and after displacement (Figure 5, bottom). Following injection of 5, there was a rapid reduction of [carbonyl-11C]6 binding in the brain, demonstrating the rapid reversibility of [carbonyl-11C]6 binding to the H3R in vivo.

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

CONCLUSION [Carbonyl-11C]6 fulfils several criteria for successful neuroreceptor imaging in vivo in non-human primates. These include: (i) high BBB permeability (ii) high specific binding and (iii) reversible binding to the H3R. The favorable kinetics of [carbonyl-11C]6 in brain may offer an improved sensitivity towards changes in endogenous histamine concentrations.

METHODS Radiochemistry. HPLC solvents were obtained from Fisher (Sweden). Unless otherwise stated, all other reagents and solvents were obtained from Sigma-Aldrich (Sweden) and used without further purification. Bromo-precursor (7) as well as the standard compounds (4-6) were supplied by AstraZeneca. General procedure for radiolabeling with [11C]CO. No-carrieradded [11C]CO2 was produced using 16.5 MeV protons in the 14 N(p,α)11C nuclear reaction on a mixture of nitrogen and oxygen gas (0.5% oxygen). [11C]CO2 was converted to [11C]CO and subsequently reacted with the coupling reagents (Bromoarene precursor (7), Pd2(π-cinnamyl)Cl2, xantphos and amine (8a-c) in anhydrous THF using a high-pressure micro-autoclave system as previously described elsewhere.28 Purification and formulation was performed using a computer controlled automated system (DMAutomation, Sweden). Semi-preparative HPLC was performed using a reversed-phase C-18 column (µBondapak, 10 µm, 10 x 300 mm, Waters) eluted with MeCN- HCO2NH4 (50 mM) 25: 75 v/v at 6 mL/min. The column outlet was connected with an absorbance detector (λ = 254 nm) in a series with radiation detector. The purified products were further purified using a solid phase extraction (SepPak, C18 plus short, Waters) and finally formulated using 5% ethanol in PBS (phosphate buffered saline, pH 7.4). Analytical chromatograms and methods for [carbonyl11 C]4-6 can be found in the supporting information.

Figure 3. Time-activity curves for the concentration of radioactivity in various brain regions as a function of time (0-123 min) following the intravenous injection of [carbonyl-11C]6 at baseline (top) and pre-treatment conditions with 5 (bottom). STR, striatum; PFC, prefrontal cortex; OC, occipital cortex; CER, cerebellum.

PET imaging in non-human primates. The study was approved by the Animal Ethics Committee of the Swedish Animal Welfare Agency (Dnr 145/08, 399/08 and 386/09) and was performed according to the “Guidelines for planning, conducting and documenting experimental research” (Dnr 4820/06-600) at the Karolinska Institutet, the Guide for the Care and Use of Laboratory Animals” the AstraZeneca bioethics policy and the EU Directive 2010/63/EU. Seven PET experiments were performed in three different experiment sessions. Session 1: Baseline PET measurements with 152157 MBq of [carbonyl-11C]4-6 in cynomolgus monkey 1 (6.9 kg). Session 2: Baseline PET measurements with 155 MBq of [carbonyl-11C]6 in cynomolgus monkey 2 (6.2 kg) followed by a pretreatment PET measurement in which AZD5213 (5, 0.1 mg/kg) was infused during 10 min starting 30 min before injection of [carbonyl-11C]6 (157 MBq). Session 3: Baseline PET measurement with 158 MBq of [carbonyl-11C]6 in cynomolgus monkey 3 (6.5 kg) followed by a displacement PET measurement in which 5 (0.1 mg/kg) was infused during 10 min starting one hour after injection of [carbonyl-11C]6 (164 MBq).

Figure 4. Representative color-coded PET-MR images showing distribution of radioactivity in cynomolgus monkey brain following the intravenous injection of [carbonyl-11C]6 at baseline (top) and pre-treatment conditions (5, 0.1 mg/kg; bottom). The images represent a summary of radioactivity from 3 to 123 min after [carbonyl-11C]6 injection. Image intensity was corrected for radioactivity.

A head fixation system was used to secure a fixed position of the monkey’s head throughout the PET measurements undertaken in each experimental session. In each PET experiment, the radiotracer was formulated in sterile physiological phosphate buffer (pH 7.4) solution containing 5 % ethanol and injected as a bolus into a sural vein during 5 s with simultaneous start of PET data acquisi-

ACS Paragon Plus Environment

Page 4 of 7

Page 5 of 7 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience tion. Radioactivity in the brain was measured continuously for 123 min according to a preprogrammed series of 34 frames.

We are grateful to Arsalan Amir, Guennadi Jogolev, Gudrun Nylén, Julio Gabriel, and all other members of the PET center at Karolinska Institutet. The authors also thank Dr. Vadim BernardGauthier for linguistic review.

REFERENCES

Figure 5. Time-activity curves for the concentration of radioactivity in various brain regions as a function of time (0-123 min) following the intravenous injection of [carbonyl-11C]6 at baseline (top) and displacement conditions with 5 (bottom). The arrow indicates the start of the infusion of 5. STR, striatum; PFC, prefrontal cortex; OC, occipital cortex; CER, cerebellum.

AUTHOR INFORMATION Corresponding Author * E-mail: [email protected]. Tel: +46-8-51775598

Author Contributions The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript.

Funding Sources The study was funded by AstraZeneca

Notes The authors declare no competing financial interest.

ACKNOWLEDGMENT

(1) Alguacil, L. F., and Perez-Garcia, C. (2003) Histamine H3 receptor: a potential drug target for the treatment of central nervous system disorders. Curr. Drug Targets: CNS Neurol. Disord. 2, 303-313. (2) Leurs, R., Vollinga, R. C., and Timmerman, H. (1995) The medicinal chemistry and therapeutic potentials of ligands of the histamine H3 receptor. Prog. Drug. Res. 45, 107-165. (3) Medhurst, A. D., Atkins, A. R., Beresford, I. J., Brackenborough, K., Briggs, M. A., Calver, A. R., Cilia, J., Cluderay, J. E., Crook, B., Davis, J. B., Davis, R. K., Davis R. P., et al. (2007) GSK189254, a novel H3 receptor antagonist that binds to histamine H3 receptors in Alzheimer’s disease brain and improves cognitive performance in preclinical models. J. Pharmacol. Exp. Ther. No. 321, 1032-1045. (4) Medhurst, A. D., Roberts, J. C., Lee, J., Chen, C. P., Brown, S. H., Roman, S., and Lai, M. K. (2009) Characterization of histamine H3 receptor in Alzheimer’s Disease brain and amyloid over-expressing TASTPM mice. Br. J. Pharmacol. 157, 130-138. (5) Narayanaswami, V., and Dwoskin, L. P. (2017) Obesity: Current and potential pharmacotherapeutics and targets. Pharmacol. Ther. 170, 116-147. (6) Phelps, M. E. (1991) PET: a biological imaging technique. Neurochem. Res. 16, 929-940. (7) Halldin, C., Gulyas, B., Langer, O., and Farde, L. (2001) PET radioligands – state of the art and new trends. J. Nucl. Med. 45, 139-152. (8) Ametamey, S. M., Honer, H., and Schubiger, P. A. (2008) Molecular imaging with PET. Chem. Rev. 108, 1501-1516. (9) Miller, P. W., Long, N. J., Vilar, R., and Gee, A. D. (2008) Synthesis of 11C, 18F, 15O, and 13N radiolabels for positron emission tomography. Angew. Chem. Int. Ed. 47, 8998-9033. (10) Windhorst, A. D., Timmerman, H., Klok, R. P., Menge, W. M., Leurs, R., and Herscheid, J. D. (1999) Evaluation of [18F]VUF5000 as a potential PET ligand for brain imaging of the histamine H3 receptor. Bioorg. Med. Chem. 7, 1761-1767. (11) Iwata, R., Horváth, G., Pascali, C., Bogni, A., Yanai, K., Kovács, Z., and Ido, T. (2000) Synthesis of 3-[1H-imidazol-4yl]propyl 4-[18F]fluorobenzyl ether ([18F]fluoroproxyfan): A potential radioligand for imaging histamine H3 reseptor. J. Label. Compd. Radiopharm. 43, 873-882. (12) Airaksinen, A. J., Jablonowski, J. A., van der Mey, M., Barbier, A. J., Klok, R. P., Verbeek, J., Herscheid, J. D. M., Leysen, J. E., Carruthers, N. I., Lammertsma, A. A., and Windhorst, A. D. (2006) Radiosynthesis and biodistribution of a histamine H3 receptor antagonist 4-[3-(4-piperidin-1-yl-but-1ynyl)-[11C]benzyl]-morpholine: evaluation of an potential PET ligand. Nucl. Med. Biol. 33, 801-810. (13) Plisson, C., Gunn, R. N., Cunningham, V. J., Bender, B., Salinas, C. A., Medhurst, A. D., Roberts, J. C., Laruelle, M., and Gee, A. D. (2009) 11C-GSK189254: A Selective Radioligand fo In Vivo Central Nervous System Imaging of Histamine H3 Receptors by PET. J. Nucl. Med. 50, 2064-2072. (14) Hamill, T. G., Sato, N., Jitsuoka, M., Tokita, S., Eng, W., Ryan, C., Krause, S., Takenago, N., Patel, S., Zeng, Z., Williams, D., Jr., Sur, C., Hargreaves, R., and Burns, H. D. (2009) Inverse agonist histamine H3 receptor PET tracer labelled with carbon-11 or fluorine-18. Synapse. 63, 1122-1132. (15) Bao, X., Lu, S., Liow, J., Zoghbi, S. S., Jenko, K. J., Clark, D. T., Gladding, R. L., Innis, R. B., and Pike, V. W. (2012) Radiosynthesis and evaluation of an 18F-Labeled Positron Emission Tomography (PET) Radioligand for Brain Histamine Subtype-3 Receptor Based on a Nonimidazole 2-

ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(16)

(17)

(18)

(19)

(20)

(21)

(22)

(23)

(24) (25)

Aminoethylbenzofuran Chemotype. J. Med. Chem. 55, 24062415. Van Laere, K. J., Sanabria-Bohórquez, S. M., Mozley, D. P., Burns, D. H., Hamill, T. G., Van Hecken, A., De Lepeleire, I., Koole, M., Bormans, G., de Hoon, J., Depré, M., Cerchio, K., Plalcza, J., Han, L., Hargreaves, R. J., and Innone, R. (2014) 11 C-MK-8278 PET as a Tool for Pharmacodynamic Brain Occupancy of Histamine 3 Receptor Inverse Agonists. J. Nucl. Med. 55, 65-72. Koga, K., Maeda, J., Tokunaga, M., Hanyu, M., Kawamura, K., Ohmichi, M., Nakamura, T., Nagai, Y., Seki, C., Kimura, Y., Minamimoto, T., Zhang, M., Fukumura, T., Suhara, T., and Higuchi, M. (2016) Development of TASP0410457 (TASP457), novel dihydroquinolinone derivative as a PETradioligand for central histamine H3 receptors. EJNMMI Research. 6, 11-25. Siméon, F. G., Culligan, W. J., Lu, S., and Pike, V. W. (2017) 11 C-Labeling of Aryl Ketones as Candidate Histamine Syptype-3 Receptor PET Radioligands through Pd(0)Mediated 11C-Carbonylative Coupling. Molecules. 22, 792802. Ashworth, S., Rabiner, E. A., Gunn, R.N., Plisson, C., Wilson, A. A., Comley, R. A., Lai, R. Y. K., Gee, A. D., Laruelle, M., and Cunningham, V. J. (2010) Evaluation of 11CGSK189254 as a Novel Radioligand for the H3 Receptor in Humans Using PET. J. Nucl. Med. 55, 1021-1029. Kimura, Y., Seki, C., Ikoma, Y., Ichise, M., Kawamura, K., Takahata, K., Moriguchi, S., Nagashima, T., Ishii, T., Kitamura, S., Niwa, F., Endo, H., Yamada, M., Higuchi, M., Zhang, M. R., and Suhara, T. (2016) [11C]TASP457, a novel PET ligand for histamine H3 receptors in human brain. Eur. J. Nucl. Med. Mol. Imging. 43, 1653-1663. Schou, M., Varnäs, K., Jureus, A., Ahlgren, C., Malmquist, J., Häggkvist, J., Tari, L., Wesolowski, S. S., Throner, S. R., Brown, D. G., Nilsson, M., Johnström, P., Finnema, S. J., Nakao, R., Amini, N., Takano, A., and Farde, L. (2016) Discovery and Preclinical Validation of [11C]AZ13153556, a Novel Probe for the Histamine Subtype 3 Receptor. ACS. Chem. Neurosci. 7, 177-184. Långström, B., Itsenko, O., and Rahman, O. (2007) [11C]Carbon monoxide, a versatile and useful precursor in labelling chemistry for PET-ligand development. J. Label. Compd. Radiopharm. 50, 794-810. Kealey, S., Gee, A. D., and Miller, P. W. (2014) Transitional metal mediated [11C]carbonylation reactions: recent advances and applications. J. Label. Compd. Radiopharm. 57, 195-201. Rahman, O. (2015) [11C]Carbon monoxide in labeling chemistry and positron emission tomography development: scope and limitations. J. Label. Compd. Radiopharm. 58, 86-98. Rotstein, B., Liang, S. H., Plazcek, M., Hooker, J. M., Gee, A. D., Dollé, F., Wilson, A. A., and Vasdev, N. (2016) 11CO bond made easy for positron emission tomography. Chem. Soc. Rev. 45, 4708-4726.

(26) Dahl, K., Halldin, C., and Schou, M. (2017) New Methodologies for the Preparation of Carbon-11 Labelled Radiopharmaceuticals. Clin. Transl. Imaging. 5, 275-289. (27) Zeisler, S. K., Nader, M., and Oberdorfer, F. (1997) Conversion of non-carrier-added [11C]carbon dioxide to [11C]carbon monoxide on molybdenum for the synthesis of 11C-labelled aromatic ketones. Appl. Radiat. Isot. 48, 1091-1095. (28) Dahl, K., Itsenko, O., Rahman, O., Ulin, J., Sjöberg, C., Sandblom, P., Larsson, L., Schou, M., and Halldin, C. (2015) An evaluation of a high-pressure 11CO carbonylation apparatus. J. Label. Compd. Radiopharm. 58, 220-225. (29) Dahl, K., Ulin, J., Schou, M., and Halldin, C. (2017) Reduction of [11C]CO2 to [11C]CO using solid supported zinc. J. Label. Compd. Radiopharm. X, XXX-XXX. (30) Dahl, K., Schou, M., Amini, N., and Halldin, C. (2013) Palladium-Mediated [11C]Carbonylation at Atmospheric Pressure: A General Method Using Xantphos as the Supporting Ligand. Eur. J. Org. Chem. 1228-1231. (31) Dahl, K., Schou, M., Rahman, O., and Halldin. C. (2014) Improved Yields for the Palladium-Mediated 11C-Carbonylation Reaction Using Microwave Technology. Clin. Transl. Imaging. 307-310. (32) Kihlberg, T., and Långström, B. (1999) Biologically active amides using palladium-mediated reactions with aryl halide and [11C]carbon monoxide. J. Org. Chem. 64, 9201-9205. (33) Pillot, C., Heron, A., Cochois, V., Tradivel-Lacombe, J., Ligneau, X., Schwartz, J. P., and Arrang, J. M. (2002)A detail mappingof the histamine H3 receptor and its gene in rat brain. Neuroscience. 114, 173-193. (34) Anichtchik, O. V., Peitsaro, N., Rinne, J. O., Kalino, H, and Panula, P. (2001) Distribution and modulation of histamine H3 receptors in basal ganglia and frontal cortex of healthy controls and patients with Parkinson’s disease. Neurobiol. Dis. 8, 707-719. (35) Jucaite, A., Takano, A., Boström, E., Jostell, K., Stenkrona, P., Halldin, C., Segerdahl, M., and Nyberg, S. (2013) AZD5213: a novel histamine H3 receptor antagonist permitting high daytime and low nocturnal H3 receptor occupancy, a PET study in human subjects. Int. J. Neuropsychopharmacol. 16, 1231-1239.

ACS Paragon Plus Environment

Page 6 of 7

Page 7 of 7 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience Authors are required to submit a graphic entry for the Table of Contents (TOC) that, in conjunction with the manuscript title, should give the reader a representative idea of one of the following: A key structure, reaction, equation, concept, or theorem, etc., that is discussed in the manuscript. Consult the journal’s Instructions for Authors for TOC graphic specifications.

Insert Table of Contents artwork here

ACS Paragon Plus Environment

7