Elsevier

NeuroImage

Volume 118, September 2015, Pages 199-208
NeuroImage

First-in-human PET quantification study of cerebral α4β2* nicotinic acetylcholine receptors using the novel specific radioligand (−)-[18F]Flubatine

https://doi.org/10.1016/j.neuroimage.2015.05.065Get rights and content

Highlights

  • Metabolization and plasma protein binding of (−)-[18F]-Flubatine are very low.

  • In cortical regions the distribution volume VT can be estimated from only 30 min PET data.

  • VT values correlated well with the known brain distribution of α4β2* nAChRs in the brain.

  • Kinetics is fast and can be well described by a 1-tissue compartment model.

  • We examined 12 healthy subjects and no adverse effects occurred.

Abstract

α4β2* nicotinic receptors (α4β2* nAChRs) could provide a biomarker in neuropsychiatric disorders (e.g., Alzheimer's and Parkinson's diseases, depressive disorders, and nicotine addiction). However, there is a lack of α4β2* nAChR specific PET radioligands with kinetics fast enough to enable quantification of nAChR within a reasonable time frame. Following on from promising preclinical results, the aim of the present study was to evaluate for the first time in humans the novel PET radioligand (−)-[18F]Flubatine, formerly known as (−)-[18F]NCFHEB, as a tool for α4β2* nAChR imaging and in vivo quantification.

Dynamic PET emission recordings lasting 270 min were acquired on an ECAT EXACT HR + scanner in 12 healthy male non-smoking subjects (71.0 ± 5.0 years) following the intravenous injection of 353.7 ± 9.4 MBq of (−)-[18F]Flubatine. Individual magnetic resonance imaging (MRI) was performed for co-registration. PET frames were motion-corrected, before the kinetics in 29 brain regions were characterized using 1- and 2-tissue compartment models (1TCM, 2TCM). Given the low amounts of metabolite present in plasma, we tested arterial input functions with and without metabolite corrections. In addition, pixel-based graphical analysis (Logan plot) was used. The model's goodness of fit, with and without metabolite correction was assessed by Akaike's information criterion. Model parameters of interest were the total distribution volume VT (mL/cm3), and the binding potential BPND relative to the corpus callosum, which served as a reference region.

The tracer proved to have high stability in vivo, with 90% of the plasma radioactivity remaining as untransformed parent compound at 90 min, fast brain kinetics with rapid uptake and equilibration between free and receptor-bound tracer. Adequate fits of brain TACs were obtained with the 1TCM. VT could be reliably estimated within 90 min for all regions investigated, and within 30 min for low-binding regions such as the cerebral cortex.

The rank order of VT by region corresponded well with the known distribution of α4β2* receptors (VT [thalamus] 27.4 ± 3.8, VT [putamen] 12.7 ± 0.9, VT [frontal cortex] 10.0 ± 0.8, and VT [corpus callosum] 6.3 ± 0.8). The BPND, which is a parameter of α4β2* nAChR availability, was 3.41 ± 0.79 for the thalamus, 1.04 ± 0.25 for the putamen and 0.61 ± 0.23 for the frontal cortex, indicating high specific tracer binding. Use of the arterial input function without metabolite correction resulted in a 10% underestimation in VT, and was without important biasing effects on BPND.

Altogether, kinetics and imaging properties of (−)-[18F]Flubatine appear favorable and suggest that (−)-[18F]Flubatine is a very suitable and clinically applicable PET tracer for in vivo imaging of α4β2* nAChRs in neuropsychiatric disorders.

Introduction

Neuronal nicotinic receptors (nAChRs) are ligand-gated ion channels that are physiologically activated by acetylcholine. Each receptor is composed of five cylindrical transmembrane subunits forming the central ion channel, and can be composed of α7 subunits or some stoichiometric combination of α and β subunits (α2–10, β2–4). The various forms are widely expressed throughout the brain. In the human brain, the most abundant form is the α4β2* subtype (Dani and Bertrand, 2007). The α4β2* nAChRs play an important role in normal brain function regulating a variety of brain processes such as mood, cognition and motor control (Changeux and Edelstein, 2005). Pathological alterations of the α4β2* nAChRs are thought to contribute to several psychiatric and neurological disorders (Dani and Bertrand, 2007, Mihailescu and Drucker-Colín, 2000, Perry et al., 1995, Rinne et al., 1991).

Noninvasive imaging using positron emission tomography (PET) or single-photon emission computer tomography (SPECT) has proved essential in the quantitative assessment of in vivo pathological changes of the α4β2* nAChR availability in neuropsychiatric disorders. High-affinity α4β2* nAChR specific radioligands, such as 5-[123I]IA-85380 (5-IA) for SPECT and 6-[18F]FA-85380 (6-FA) and 2-[18F]FA-85380 (2-FA) for PET, have been developed and optimized for α4β2* nAChR imaging (Chefer et al., 1998, Ding et al., 2004, Doll et al., 1999, Horti et al., 1998, Kimes et al., 2003, Mamede et al., 2004, Scheffel et al., 2000, Schildan et al., 2007). Recently, through the use of 2-FA-PET and 5-IA-SPECT, cortical and subcortical abnormalities of the α4β2* nAChR binding have been reported in a variety of neuropsychiatric disorders such as Alzheimer's disease (AD) and mild cognitive impairment (MCI) (Kendziorra et al., 2011, O'Brien et al., 2007, Okada et al., 2013, Sabri et al., 2008), Parkinson's disease (PD) (Fujita et al., 2006, Kas et al., 2009, Meyer et al., 2009), epilepsy (Picard et al., 2006), nicotine dependence (Mukhin et al., 2008, Staley et al., 2006), major depressive disease (Saricicek et al., 2012), and schizophrenia (D'Souza et al., 2012, Brašić et al., 2012).

However, the slow kinetics of the PET radioligand 2-[18F]FA-85380 and SPECT radioligand 5-[123I]IA-85380 require lengthy acquisition times of up to 7 h to enable whole-brain nAChR analysis. Therefore, the use of 2-FA or 5-IA for large-scale clinical trials and routine clinical application may be limited (Horti and Villemagne, 2006, Horti et al., 2010, Sabri et al., 2008). A new generation α4β2* nAChR specific PET radioligands, such as (−)-[18F]norchloro-fluoro-homoepibatidine ((−)-[18F]NCFHEB), also known as (−)-[18F]Flubatine, [18F]AZAN and [18F]nifene, demonstrating faster kinetic properties in both preclinical and in-man investigations have been developed (Brust et al., 2008, Hillmer et al., 2011, Hockley et al., 2013, Kuwabara et al., 2012, Sabri et al., 2008, Wong et al., 2013). We recently developed (−)-[18F]Flubatine as a less toxic derivative of epibatidine (Bai et al., 1996, Brust et al., 2008, Deuther-Conrad et al., 2004, Deuther-Conrad et al., 2008, Fischer et al., 2013, Patt et al., 2003, Patt et al., 2014, Patt et al., 2013, Sabri et al., 2008, Smits et al., 2014). Preclinical studies demonstrated that (−)-[18F]Flubatine has high selectivity and specificity for α4β2* nAChRs (Brust et al., 2008, Deuther-Conrad et al., 2004, Smits et al., 2014).

The objectives of the present study were to determine the safety and tolerability of (−)-[18F]Flubatine, and to evaluate kinetic model-based approaches to quantify α4β2* nAChR binding parameters from dynamic PET and blood data in healthy volunteers.

Section snippets

Human subjects

Twelve healthy male volunteers (age: 71.0 ± 5.0 years, range: 63 to 76 years) took part in the study. All were non-smokers, drug free for any kind of centrally acting medication, and had no history of neurological or psychiatric illness. They were recruited by newspaper advertisement and from an existing database at the Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig. All study subjects underwent a thorough clinical assessment, and were required to achieve results within one

Blood plasma and kinetic analyses

In blood plasma, almost 90% of unchanged (−)-[18F]Flubatine as measured by radio-HPLC was found at 90 min and 85% at 270 min p.i. (Patt et al., 2014). This demonstrates that (−)-[18F]Flubatine is very stable (Fig. 2). Notably, in an animal study (juvenile female pigs) we measured about 60% metabolites in plasma at 90 min after injection of (−)-[18F]Flubatine (Brust et al., 2008). Please note that in this reference the older name (−)-[18F]NCFHEB is used. Plasma protein binding was 15.2 ± 1.0%.

Discussion

This study describes the first-in-human application of a new PET tracer (−)-[18F]Flubatine, developed as a viable alternative to the slow tracer kinetic PET and SPECT radioligands 2-[18F]FA-85380 and 5-[123I]IA-85380. (−)-[18F]Flubatine is a less toxic derivative of epibatidine with favorable preclinical results in terms of α4β2* nAChR specificity and selectivity (Brust et al., 2008, Deuther-Conrad et al., 2004, Deuther-Conrad et al., 2008, Gallezot et al., 2014, Hockley et al., 2013).

Several

Conclusion

In this study we have shown that (−)-[18F]Flubatine can produce quantitative PET measurements of α4β2* nAChR availability in the human brain. It offers several advantages. Because metabolization of the tracer is small, it is not necessary to correct the arterial input function for metabolites. Binding to plasma proteins is also of minor importance, and because the free fraction of (−)-[18F]Flubatine in plasma is uniformly high, a correction of VT for fP can also be omitted. The kinetics of (−)-[

Acknowledgments

The study was supported by a BMBF grant (German Federal Ministry of Education and Research; 01EZ0820, 01EZ0821, 01EZ0822 and 01EZ0823). S.G. received funding from the International Max Planck Research School on Neuroscience of Communication (IMPRS NeuroCom). We wish to thank all volunteers who participated in this trial. We are also grateful to the cyclotron, PET radiochemistry, and PET scanner crews of the Leipzig University Department of Nuclear Medicine for their skillful support. We are

References (57)

  • M. Patt et al.

    Evaluation of metabolism, plasma protein binding and other biological parameters after administration of (−)-[18F]Flubatine in humans

    Nucl. Med. Biol.

    (2014)
  • E.K. Perry et al.

    Alteration in nicotine binding sites in Parkinson's disease, Lewy body dementia and Alzheimer's disease: possible index of early neuropathology

    Neuroscience

    (1995)
  • U. Scheffel et al.

    6-[18F]Fluoro-A-85380: an in vivo tracer for the nicotinic acetylcholine receptor

    Nucl. Med. Biol.

    (2000)
  • A. Schildan et al.

    Synthesis procedure for routine production of 2-[18F]fluoro-3-(2(S)-azetidinylmethoxy)pyridine (2-[18F]F-A-85380)

    Appl. Radiat. Isot.

    (2007)
  • R. Smits et al.

    Synthesis and biological evaluation of both enantiomers of [(18)F]flubatine, promising radiotracers with fast kinetics for the imaging of α4β2-nicotinic acetylcholine receptors

    Bioorg. Med. Chem.

    (2014)
  • D. Sorger et al.

    Binding properties of the cerebral alpha4beta2 nicotinic acetylcholine receptor ligand 2-[18F]fluoro-A-85380 to plasma proteins

    Nucl. Med. Biol.

    (2006)
  • D. Bai et al.

    Synthesis of (+/−)-epibatidine and its analogues

    J. Org. Chem.

    (1996)
  • G. Becker et al.

    PET quantification of (−)-[F-18]-flubatine binding to nicotinic alpha4beta2 acetylcholine receptors in human brains

    J. Cereb. Blood Flow Metab.

    (2012)
  • J.R. Brašić et al.

    Positron emission tomography experience with 2-[18F]fluoro-3-(2(S)-azetidinylmethoxy)pyridine (2-[18F]FA) in the living human brain of smokers with paranoid schizophrenia

    Synapse

    (2012)
  • A.L. Brody et al.

    Cigarette smoking saturates brain α4β2 nicotinic acetylcholine receptors

    Arch. Gen. Psychiatry

    (2006)
  • P. Brust et al.

    In vivo measurement of nicotinic acetylcholine receptors with [18F]norchloro-fluoro-homoepibatidine

    Synapse

    (2008)
  • K.P. Burnham et al.

    Model Selection and Multimodel Inference: A Practical Information Theoretic Approach

    (2002)
  • J.-P. Changeux et al.

    Nicotinic Acetylcholine Receptors

    (2005)
  • S.I. Chefer et al.

    In vivo imaging of brain nicotinic acetylcholine receptors with 5-[123I]iodo-A-85380 using single photon emission computed tomography

    Life Sci.

    (1998)
  • J.A. Dani et al.

    Nicotinic acetylcholine receptors and nicotinic cholinergic mechanisms of the central nervous system

    Annu. Rev. Pharmacol. Toxicol.

    (2007)
  • Y.S. Ding et al.

    6-[18F]Fluoro-A-85380, a new PET tracer for the nicotinic acetylcholine receptor: studies in the human brain and in vivo demonstration of specific binding in white matter

    Synapse

    (2004)
  • F. Doll et al.

    Synthesis and nicotinic acetylcholine receptor in vivo binding properties of 2-fluoro-3-[2(S)-2-azetidinylmethoxy]pyridine: a new positron emission tomography ligand for nicotinic receptors

    J. Med. Chem.

    (1999)
  • D.C. D'Souza et al.

    Lower ß2*-nicotinic acetylcholine receptor availability in smokers with schizophrenia

    Am. J. Psychiatry

    (2012)
  • Cited by (48)

    • Inconsistencies in atlas-based volumetric measures of the human nucleus basalis of Meynert: A need for high-resolution alternatives

      2022, NeuroImage
      Citation Excerpt :

      In addition, ACh has a well-established role in modulating experience-dependant synaptic changes, and hence in learning and memory (Ang et al., 2021; Easton et al., 2012; Hasselmo 1999, 2006; Mitsushima et al., 2013; Obermayer et al., 2017; Picciotto et al., 2012). Moreover, cholinergic dysfunction has also been considered as a key factor in neurodegenerative diseases such as Alzheimer's disease (AD) (Arendt et al., 1983; Hanna Al-Shaikh et al. 2020; McGeer et al., 1984; Nagai et al., 1983; Rinne et al., 2003; Sabri et al., 2015; Vogels et al., 1990; Whitehouse et al., 1981; Wu et al., 2010) and Parkinson's disease (Arendt et al., 1983; Bohnen et al., 2012, 2015; Chan‐Palay 1988; Gaspar and Gray 1984; Hall et al., 2014; Klein et al., 2010; Kuhl et al., 1996; Müller et al., 2015; Nakano and Hirano 1984; Tagliavini et al., 1984; Whitehouse et al., 1983; Zarow et al., 2003) and in neurodevelopmental disorders such as Schizophrenia (Carruthers et al., 2015). With such significant roles in neuronal function, cognition and neurodegenerative diseases, the cholinergic pathway, and especially the nbM, is of particular interest to both clinical and fundamental neuroscience.

    View all citing articles on Scopus

    The asterisk applied in the receptor nomenclature notifies that the receptor complex may include additional subunits.

    1

    Contributed equally.

    View full text