ArticlePDF Available

Radiosynthesis and first preclinical evaluation of the novel norepinephrine transporter pet-ligand [11C]ME@HAPTHI

Authors:

Abstract and Figures

The norepinephrine transporter (NET) has been demonstrated to be relevant to a multitude of neurological, psychiatric and cardiovascular pathologies. Due to the wide range of possible applications for PET imaging of the NET together with the limitations of currently available radioligands, novel PET tracers for imaging of the cerebral NET with improved pharmacological and pharmacodynamic properties are needed. The present study addresses the radiosynthesis and first preclinical evaluation of the novel NET PET tracer [(11)C]Me@HAPTHI by describing its affinity, selectivity, metabolic stability, plasma free fraction, blood-brain barrier (BBB) penetration and binding behaviour in in vitro autoradiography. [(11)C]Me@HAPTHI was prepared and displayed outstanding affinity and selectivity as well as excellent in vitro metabolic stability, and it is likely to penetrate the BBB. Moreover, selective NET binding in in vitro autoradiography was observed in human brain and rat heart tissue samples. All preclinical results and radiosynthetic key-parameters indicate that the novel benzothiadiazole dioxide-based PET tracer [(11)C]Me@HAPTHI is a feasible and improved NET radioligand and might prospectively facilitate clinical NET imaging.
Content may be subject to copyright.
O R I G I N A L R E S E A R C H Open Access
Radiosynthesis and first preclinical
evaluation of the novel norepinephrine
transporter pet-ligand [
11
C]ME@HAPTHI
Christina Rami-Mark
1,2
, Neydher Berroterán-Infante
1,2
, Cecile Philippe
1,3
, Stefanie Foltin
1
, Chrysoula Vraka
1
,
Alexander Hoepping
4
, Rupert Lanzenberger
5
, Marcus Hacker
1
, Markus Mitterhauser
1,3*
and Wolfgang Wadsak
1,2
Abstract
Background: The norepinephrine transporter (NET) has been demonstrated to be relevant to a multitude of
neurological, psychiatric and cardiovascular pathologies. Due to the wide range of possible applications for PET
imaging of the NET together with the limitations of currently available radioligands, novel PET tracers for imaging
of the cerebral NET with improved pharmacological and pharmacodynamic properties are needed.
Methods: The present study addresses the radiosynthesis and first preclinical evaluation of the novel NET PET tracer
[
11
C]Me@HAPTHI by describing its affinity, selectivity, metabolic stability, plasma free fraction, bloodbrain barrier
(BBB) penetration and binding behaviour in in vitro autoradiography.
Results: [
11
C]Me@HAPTHI was prepared and displayed outstanding affinity and selectivity as well as excellent
in vitro metabolic stability, and it is likely to penetrate the BBB. Moreover, selective NET binding in in vitro
autoradiography was observed in human brain and rat heart tissue samples.
Conclusions: All preclinical results and radiosynthetic key-parameters indicate that the novel benzothiadiazole
dioxide-based PET tracer [
11
C]Me@HAPTHI is a feasible and improved NET radioligand and might prospectively facilitate
clinical NET imaging.
Keywords: NET; PET; Autoradiography; Radiosynthesis; HAPTHI
Background
The noradrenergic systemand specifically the presynaptic
norepinephrine transporter (NET)is proposed to be
altered in a variety of neurological, neuropsychiatric
and cardiovascular diseases. For example, alterations have
been shown in Alzheimers disease, Morbus Parkinson,
major depressive disorder and attention deficit hyper-
activity disorder [19]. Therefore, a reliable non-invasive
molecular imaging techniquesuch as positron emission
tomography (PET)would be of great benefit for early
stage in vivo diagnostics, visualization of treatment response
and further elucidation of underlying pathophysiological
mechanisms.
Great efforts have been made to develop PET tracers
for the NET over the last two decades. Focus was pri-
marily placed on reboxetine-derived ligands [1014].
However, previous studies have shown that the in vivo
and in vitro behaviour of these reboxetine analogues,
more specifically [
11
C]MeNER ([
11
C]MRB, ((S,S)-2-(α-(2-
[
11
C]methoxyphenoxy)benzyl)morpholine), [
11
C]MeNET
and [
18
F]FMeNER-D
2
((S,S)-2-(α-(2-[
18
F]fluoro[
2
H
2
]
methoxyphenoxy)benzyl) morpholine), is not favourable
for viable imaging of the NET by PET. Limitations include
their metabolic stability, late reaching of equilibrium,
unexplainable striatal uptake and complexity of radiosynth-
esis [10, 1518]. Recently, we aimed at the preparation of a
benzo[d]imidazolone derivative[
11
C]Me@APPI as new
NET PET tracer [19]. Despite its favourable properties and
straightforward production, its affinity was not sufficient
* Correspondence: markus.mitterhauser@meduniwien.ac.at
Equal contributors
1
Department of Biomedical Imaging and Image-guided Therapy, Division of
Nuclear Medicine, Medical University of Vienna, Vienna, Austria
3
Faculty of Life Sciences, Department of Technology and Biopharmaceutics,
University of Vienna, Vienna, Austria
Full list of author information is available at the end of the article
© 2015 Rami-Mark et al. This is an Open Access article distributed under the terms of the Creative Commons Attribution
License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly credited.
Rami-Mark et al. EJNMMI Research (2015) 5:34
DOI 10.1186/s13550-015-0113-3
and its lipophilicity high. Hence, there is ample demand for
a novel, improved radioligand for in vivo NET imaging.
Therefore, this study highlights a novel, non-reboxetine-
based NET PET tracer based on a benzothiadiazole
scaffold: [
11
C]Me@HAPTHI ((S)-1-(3-hydroxy-4-([
11
C]
methylamino)butyl)-3-phenyl-1,3-dihydrobenzo[c][1, 2,
5]thiadiazole 2,2-dioxide) (Fig. 1). In general, the de-
signed benzothidiazole dioxides exhibits excellent affin-
ity and selectivity as well as slightly reduced flexibility
compared to other previously published benzoimidazo-
lones [20, 21]. Hence, these substances offer an ideal
basis for the further development of novel NET ligands
for PET imaging.
The objectives of this investigation were as follows:
The set-up of a small-scale radiosynthetic procedure
for the preparation of the carbon-11 labelled
[
11
C]Me@HAPTHI and its optimization;
Theup-scalingandset-upofafullyautomated
preparation of [
11
C]Me@HAPTHI, including
purification and formulation;
The in vitro evaluation of Me@HAPTHI and its
precursor HAPTHI. Evaluation includes binding
studies for the determination of affinity and
selectivity of both Me@HAPTHI and its precursor
HAPTHI towards NET using NET, serotonin
transporter (SERT) and dopamine transporter
(DAT) expressing membranes, metabolic stability
testing in vitro against Cytochrom P 450 enzymes,
logP analysis and immobilized artificial membrane
(IAM) chromatography for indirect measurement of
the bloodbrain barrier (BBB) penetration and
determination of plasma free fraction.
Comparative in vitro autoradiography on human
and rodent tissue slices.
Methods
Materials
Precursor, HAPTHI ((S)-1-(4-amino-3-hydroxybutyl)-
3-phenyl-1,3-dihydrobenzo[c][1, 2, 5]thiadiazole 2,2-
dioxide, and cold reference compound Me@HAPTHI
((S)-1-(3-hydroxy-4-(methylamino)butyl)-3-phenyl-1,3-
dihydrobenzo[c][1, 2, 5]thiadiazole 2,2-dioxide) were
custom-synthesized by ABX Advanced Biochemical
Compounds (Radeberg, Germany). Briefly, synthesis of
(2S)-4-(2,2-dioxido-3-phenyl-2,1,3-benzothiadiazol-1(3H)-
yl)-1-(methylamino)butan-2-ol followed the route de-
scribed by Neill et al. [20, 21]. For more details, see
Additional file 1.
2-Butanone (MEK, <99.0 % ACS reagent), acetonitrile
(ACN, HPLC grade), dimethylsulfoxide (DMSO), tetrabu-
tylammonium hydroxide 30-hydrate (TBAH), ammonium
formate, ammonium acetate, sodium hydroxide, triethyla-
mine and ethanol (absolute) were purchased from Sigma-
Aldrich (Vienna, Austria) in the highest available grades.
In addition, iodine (sublimated grade for analysis; ACS,
Pharm. Eur.) was obtained from Merck (Darmstadt,
Germany). Silver triflate impregnated carbon was pre-
pared by reaction of 1 g of silver trifluoromethanesulfo-
nate (Sigma Aldrich, Vienna, Austria) in 20 mL ACN with
3 g of Graphpac-GC (80/100 mesh, Alltech, Deerfield,
USA). The suspension was stirred under protection
from light and in an argon atmosphere for 30 min.
After removal of the solvent, the resulting powder was
dried under protection from light for further 2 h under re-
duced pressure.
For formulation of the product, 0.9 % saline solution
from B. Braun (Melsungen, Germany), 3 % saline solution
(Landesapotheke Salzburg, Austria) and sodium dihydro
genphosphate-monohydrate and disodiumhydrogenphos
phate-dihydrate (both from Merck, Darmstadt, Germany)
Fig. 1 Chemical structures of reboxetine and
11
C-labelled NET PET tracers [
11
C]Me@APPI and [
11
C]MeNER, [
11
C]MeNET and our novel NET PET
ligand [
11
C]ME@HAPTHI. The red coloured atom indicates the position of the radioisotope introduced by radiolabeling
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 2 of 12
were used. Sterile water was purchased from Meditrade
Medicare Medizinprodukte (Kufstein, Austria). Phosphate
buffer (125 mM) was prepared by dissolving 0.224 g so-
dium dihydrogenphosphate-monohydrate and 1.935 g
disodiumhydrogenphosphate-dihydrate in 100 mL ster-
ile water. For solid phase extraction, C18 plus SepPak®
cartridges were purchased from Waters (Waters® Asso-
ciates, Milford, USA). Low-protein binding Millex® GS
0.22 μm sterile filters were obtained from Millipore
(Bedford, USA).
All other chemicals and solvents for the radiosyntheses
were obtained from Merck (Darmstadt, Germany) and
Sigma-Aldrich (Vienna, Austria) with at least analytical
grade and used without further purification.
NET, DAT and SERT expressing membrane prepara-
tions were obtained from Perkin Elmer (MA, USA). An
ODP-50column(20×4.0mm,5μm) was purchased
from Shodex® (Showa Denko Europe GmbH, Munich,
Germany). For prediction of BBB penetration, a Redistech
IAM.PC.DD2 column (Regis Technologies Inc., Morton
Grove, USA) was used.
Microsomal preparations (human/rat liver microsomes)
for stability testing were obtained from BD Bioscience (NJ,
USA). Pooled human and rat plasma was obtained from
Innovative Research (MI, USA).
The human postmortem tissue (79hpostmortem
time, no history of neurological diseases) was obtained
from the Neurobiobank of the Medical University of
Vienna and approved by the local ethics committee
(Molecular neuropathologic investigation of neurodegen-
erative diseasesNr.396/2011) following the principles of
the Helsinki Declaration. Wild-type male rats were deeply
anesthesized by isoflurane and sacrificed by decapitation.
The organs of interest (i.e. brain, heart and testis) were re-
moved and quick-frozen in i-pentan. Research using ani-
mal tissue was carried out under institutional approval in
accordance with the Austrian Animal Care Law. Tissues
were cut at 20 °C in a micro-cryotome (Microm HM
560, Thermo Scientific, Austria). Frozen slices were
thaw-mounted onto superfrost slides (Menzel-Gläser
SUPERFROST plus microscopy slides, Thermo Scientific,
Germany). A barrier pen (Mini PAP Pen, Invitrogen,
USA) was used for immunohistochemistry only. For de-
tection of autoradiography, a Cyclone Phospho-Imager
(Cyclone Plus Storage Phosphor System, Perkin Elmer,
Germany) and Phosphor Imager plates (Multisensitive
Phosphor Screens Long Type MS, PPN 7001724, Perkin
Elmer, Germany) were used. The lead shielded and
light-protected cassettes (Fisher Biotech Autoradiog-
raphy Cassette FBCS 1417) were purchased from Fisher
Scientific (PA, USA).
TheNET-antibody(SLC6A2AntibodyH-67,sc-
67216) was purchased from Santa Cruz Biotechnology
(TX, USA). An endogenous Avidin-Biotin blocking kit
(ab64212) as well as the DAB (=3,3-diaminobenzidine)
substrate kit (94665) was obtained from abcam (Cambridge,
UK). A rabbit primary antibody isotype control was
purchased from Invitrogen (CA, USA). A peroxidase-
based Vectastain ABC kit (Rabbit IgG, PK-4001) was
obtained from Vector Laboratories (CA, USA). Phos-
phate buffered saline (PBS pH 7.4, tenfold concentrate,
11237) was obtained from Morphisto Evolutionsforschung
und Anwendung GmbH (Germany). Mayers Hemalaun
solution was purchased from Merck Millipore (Germany).
Histofluid (Marienfeld Superior, Germany) was used as a
mounting medium. Coverslips from Menzel Gläser (24 ×
60 mm, Thermo Fisher Scientific, Germany) were used for
conservation of mounted slides. All other chemicals were
obtained from Sigma-Aldrich.
Instrumentation
[
11
C]CO
2
was produced within a GE PETtrace cyclotron
(General Electric Medical System, Uppsala, Sweden) by a
14
N(p,α)
11
C nuclear reaction under irradiation of a gas
target filled with N
2
(+1 % O
2
) (Air Liquide Austria
GmbH, Schwechat, Austria).
The evaluation of the reaction conditions was performed
manually with starting activities <2 GBq. After optimization
of the reaction parameters, [
11
C]Me@HAPTHI-synthesis
was transferred to the TRACERlabFX C Pro synthesizer
and a fully automated synthesis was established.
Crude [
11
C]Me@HAPTHI was purified by semi-
preparative reversed phase HPLC using the built-in
semi-preparative HPLC system equipped with a radio-
activity and a UV detector (Linear Instruments Model
200 Detector UV/VIS) and a LaPrep HPLC pump (VWR
International, Radnor, USA). A Supelcosil
TM
LC-ABZb,
5μm, 250 × 10 mm (Supelco®, Bellefonte, PA, USA) col-
umn was used with a mobile phase of ACN/0.1 M am-
monium acetate 40/60 v/v% at a flow rate of 6 mL/min.
The analytical HPLC was performed on a Merck-Hitachi
LaChrom HPLC system (L-7100 pump; LaChrom L-7400
UV detector) using a NaI radio-detector (Bertholdt
Technologies, Bad Wildbach, Germany) and a GinaStar®
processing software (Raytest, Straubenhardt, Germany). A
Phenomenex® Prodigy, Phenyl-3(PH-3), 5 μm, 250 ×
4.6 mm (Phenomenex®, Aschaffenburg, Germany) column
with a mobile phase consisting of ACN/0.1 M ammonium
formate 50/50 v/v% at a flow rate of 2 mL/min was used
while detection of the cold compounds was performed
at 280 nm.
The osmolality of the final sterile product was mea-
sured with a Wescor osmometer Vapro® 5600 (Sanova
Medical Systems, Vienna, Austria).
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 3 of 12
Methods
Radiochemistry
Production of [
11
C]CH
3
I and [
11
C]CH
3
OTf
The cyclotron production of [
11
C]CO
2
was terminated
at desired target activities between 40 and 50 GBq at
currents between 48 and 53 μA(2025 min) and trapped
upon delivery on a molecular sieve (4 Å) within the
Tracerlab FxC Pro synthesizer. Subsequently, [
11
C]CO
2
was converted into [
11
C]CH
4
by a Ni-catalysed reduc-
tion with H
2
at 400 °C. [
11
C]CH
3
I was produced within
the same synthesizer using the dry method (gas phase
conversion) described by Larsen et al. [22] with adopted
modifications described by Kniess et al. [23]. Briefly,
the resulting [
11
C]CH
4
was reacted with sublimated
iodine at 738 °C in a recirculating process for 4 min to
give [
11
C]CH
3
I. The produced [
11
C]CH
3
Iwastrapped
on-line on a Porapak® N column and finally released by
heating the trap to 190 °C. [
11
C]CH
3
OTf was prepared
on-line at the passage of [
11
C]CH
3
Ithroughapre-heated
(200 °C) column containing 300 mg silver triflate impreg-
nated graphitized carbon at a flow rate of 40 mL/min [24].
Small-scale reactions
For optimization of reaction conditions, small-scale reac-
tions using [
11
C]CH
3
Ior[
11
C]CH
3
OTf were performed.
Either [
11
C]CH
3
Ior[
11
C]CH
3
OTf was trapped in 500 μL
of the solvent of choice at room temperature (RT) and
aportioned for further experiments in 1 mL Wheaton v-
vials. All evaluation reactions were performed manually
(shielded hood; starting activity <2 GBq). The influence of
various reaction conditions was investigated:
Reaction temperature: 25 °C, 75 °C
Base as catalyst: NaOH, triethylamine (TEA) and
TBAH
Precursor concentration: 1 or 2 mg/mL
Solvent: MEK or DMSO
Finale reaction volumes of small-scale reactions were
10200 μL. The reactions were quenched with an equi-
volume solution of ammonium acetate (aq., pH 3.5), and
the radiochemical yield (RCY) was determined using
analytical radio-HPLC. In Fig. 2, the reaction scheme is
presented.
Full automation of radiosyntheses
The automation of the N-
11
C-methylation reaction was
done on the TRACERlab
TM
FX C Pro (GE Healthcare).
A schematic flowchart of the synthesis is depicted in
Fig. 3.
After conversion of cyclotron-produced [
11
C]CO
2
to
[
11
C]methane, [
11
C]methyl iodide and [
11
C]CH
3
OTf, it
was trapped at RT in a glass reactor containing precur-
sor HAPTHI (1 mg, 3 μmol) and 0.5 μL of an aqueous
NaOH-solution (5 M) in 500 μL MEK. After heating of
the sealed reaction vessel to 75 °C for 2 min, the crude
reaction mixture was cooled to 25° and quenched by
addition of 1 mL HPLC eluent. The entire volume was
then transferred to the 5 mL injection loop. The crude
mixture was (fluid detector controlled) injected into the
semi-preparative HPLC column (Fig. 4). The pure
[
11
C]Me@HAPTHI peak was cut into a round bulb, con-
taining 80 mL of distilled water. The now predominantly
Fig. 2 Radiosynthesis of [
11
C]Me@HAPTHI starting from the precursor molecule HAPTHI
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 4 of 12
aqueous product solution was subjected to solid phase
extraction by transferring over a preconditioned (10 mL
EtOH, air, 20 mL water) C18 SPE cartridge. After rinsing
of the C18 SepPak® with water (V6) for complete re-
moval of residual HPLC solvents, the pure product was
eluted with 1.5 mL EtOH (V5) into a two-neck vial and
the cartridge and transfer lines rinsed with further 5 mL
0.9 % saline into the same vial. After formulation with
9 mL 0.9 % saline, 1 mL 3 % saline and 1 mL 125 mM
phosphate buffer, sterile filtration (0.22 μm) was performed
Fig. 3 Flow scheme of the fully automated radiosynthesis of [
11
C]Me@HAPTHI
Fig. 4 aSemi-preparative and banalytical HPLC chromatogram
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 5 of 12
under aseptic conditions (laminar air flow hot cell, class A)
to avoid microbial contamination.
Quality control
Chemical and radiochemical impurities were assessed
using analytical radio- and UV-HPLC according to the
monograph in the European Pharmacopoeia [25]. Radio-
chemical identity and purity were measured via analyt-
ical radio-HPLC by comparison of retention times with
authentic samples. Specific radioactivity was determined
by quantification of the non-radioactive product (HPLC
UV channel at 280 nm) and inclusion of the overall
radiochemical yield (GBq at end of synthesis). Sterility,
absence of endotoxins, pH, osmolality and residual sol-
vents were determined by standard procedures routinely
performed at the PET Centre of the Vienna General
Hospital/Medical University of Vienna and follow the re-
spective monograph in the European Pharmacopoeia [25].
Statistical analysis
All quantitative data described in the text and figures
are specified as arithmetic mean ± standard deviation.
For the determination of significance, a Students two-
tailed ttest (α= 0.95) was performed using Microsoft®
Excel. Pvalues of <0.05 were considered to be signifi-
cant. Unless otherwise stated, error bars in figures repre-
sent the standard deviation; if not visual, they are within
the icon margin.
NET-expressing membrane binding studies
The affinity of new radiolabelled ligand was determined
in a NET-expressing membrane binding protocol [26, 27].
For details, see Additional file 1.
Data from the competition plots (as arithmetic means
of values derived from three different assays, each in
triplicate for each compound) were analyzed and subse-
quently IC
50
and K
i
values were calculated using GraphPad
Prism® software (San Diego, USA).
Assays similar to those described for NET were per-
formed in order to determine the selectivity of the
tested compounds towards NET in comparison to DAT
and SERT. IC
50
and K
i
values were obtained in analogy
to NET experiments. Ratios DAT/NET and SERT/NET
were determined.
LogD analysis, IAM chromatography and bloodbrain
barrier penetration
LogD values were assessed using a HPLC-based protocol
according to Donovan and Pescatore [28]. All compounds
(as cold reference standards) were injected together
with two known compoundswith known logD and k
valuesaccording to a standard protocol. A polymeric
ODP-50 column was used; a linear gradient from 10 %
MeOH 90 % 25 mM phosphate buffer (pH 7.4) to
100%methanolwithin9.4minataflow-rateof
1.5 mL/min was applied. Internal standards were tri-
phenylene and toluene; detection was performed at 260
and 285 nm.
As lipophilicity alone was shown to be a tenuous pre-
dictor for bloodbrain barrier penetration, other in vitro
methods have been described, such as immobilized artifi-
cial membrane (IAM) chromatography and further calcu-
lation of total polar surface area (tPSA) values [2931].
Therefore, IAM chromatography was performed using a
Redistech IAM.PC.DD2 column (15 cm × 4.6 mm) ac-
cording to previously published methods [19, 3235]. For
analysis, 0.01 M phosphate buffer (pH 7.4) and ACN (in
different ratios) were used isocratically as mobile phase
at a flow rate of 1 mL/min. Resulting K
m
(membrane
partition coefficient) and P
m
(permeability) values were
obtained after data analysis using Microsoft Excel. The
resulting data were compared with those derived from
compounds known to penetrate BBB as external stand-
ard. Additionally, tPSA values were determined in silico
using Chem Bio Draw Ultra (Cambridge Software, Perkin
Elmer, USA).
Metabolic stability testing
Pooled human and rat liver microsomes are subcellular
fractions that are rich in endoplasmatic reticuli, which
contain many drug-metabolizing enzymes, e.g. cytochrome
P450s, flavin monooxygenases and epoxide hydrolase.
Microsomal incubations were performed in order to in-
vestigate the metabolization of [
11
C]Me@HAPTHI. As
the results, both the percentage of test compound me-
tabolized after a certain time and the biological half-life
were determined.
Plasma protein binding
For the determination of free fraction in human pooled
plasma, an ultrafiltration protocol according to previ-
ously published methods was used [3538]. Briefly,
aliquots of pooled human plasma were spiked with
[
11
C]Me@HAPTHI and centrifuged using ultrafiltra-
tion vials (Amicon Centrifree; Millipore, Bedford,
USA). The plasma free fraction was calculated, and the
percentage of unspecific binding of [
11
C]Me@HAPTHI to
the filter matrix evaluated. For a detailed method, see
Additional file 1.
Autoradiography, Nissl staining and
immunohistochemistry
Human brain tissue (cortex, thalamus, hippocampus,
cerebellum and hypothalamus) was obtained deeply
frozen from the Neurobiobank of the Medical University
Vienna and was stored at 80 °C. Before cutting, tissue
blocks were thawed slowly within 12 h to 20 °C. The
organs were cut at 20 °C in a micro-cryotome into 10-
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 6 of 12
μm-thick slices and thaw mounted onto object slides.
Slices were again stored at 80 °C until the beginning
of the experiment.
In vitro autoradiography was performed with slight
modifications according to previously published protocols
[13, 39, 40]. Non-specific binding was determined by
co-incubation with excess Nisoxetine (10 μM). For
competition experiments, non-radioactive FMeNER-D2,
an established NET PET tracer, and Me@HAPTHI were
added to the incubation solution in different concen-
trations. After 1 h at room temperature, incubation
was stopped and slices were processed on phosphor
imaging films.
All data was exported to Microsoft Excel for statistical
analysis, and the percentage of total specific binding was
calculated.
Post-autoradiographic processing of the slices was done
by Nissl staining in order to facilitate morphological map-
ping of hot areas in the autoradiography. The same tissue
slices were stained after autoradiography with cresyl violet
[28, 41, 42] to demonstrate the Nissl substance in the
neurons and cell nuclei. For a detailed procedure, see
Additional file 1.
Immunohistochemical staining experiments were per-
formed on rat and human tissue cryo-slices, vicinal to the
slices used for autoradiographic experiments. The staining
procedure was a modification of a general protocol as
published previously in detail [28, 43].
Results
Radiochemistry
The optimum parameters were examined in small-scale
reactions. Thus, the influence of different
11
C-methyla-
tion agents, solvent, precursor concentration, reaction
temperature and base were investigated (Fig. 5ad).
Radiochemical yields (RCY) of [
11
C]Me@HATPHI were
below 6 % for all examined conditions using [
11
C]CH
3
I
as methylation agent. Hereby, DMSO proved to be the
best solvent for the SN
2
reaction using [
11
C]methyl iod-
ide. In contrast, very promising results were obtained
using [
11
C]CH
3
OTf as radio-methylation agent (Fig. 5cd).
Interestingly, the use of DMSO as solvent did not result in
high yields, less than 1 % RCY was observed using
[
11
C]CH
3
OTf. Applying 2-butanone resulted in high
radiochemical yields. Furthermore, the influence of
basic catalysis was examined: TBAH catalysis could not
shift the reaction kinetics to favourable outcomes, as it
did not result in any methylation of HAPTHI. Up to
12.8 ± 4.7 % RCY was observed when using 0.5 μL
triethylamine instead. Conducting the experiments with
0.5 μL of 1 M NaOH (aq.), however, yielded 42.9 ±
5.2 % radiochemical yield with 1 mg/mL precursor con-
centration and even above 50 % RCY were obtained
with 2 mg/mL precursor concentration. A further in-
crease in basicityfacilitated by 0.5 μL5MNaOH
(aq.) instead of 1 M NaOH (aq.)did not lead to im-
proved results (in a total reaction volume of 100 μL);
only <0.5 % RCY were obtained.
Hence, the best results were obtained with NaOH-
catalysis in 2-butanone for 2 min at 75 °C using 2 mg/
mL precursor HAPTHI. Thereby, 54.0 ± 8.3 % radio-
chemical yield was achieved.
Therefore, these optimum reaction parameters were
transferred to the fully automated radiosynthesis within
the Tracerlab FxC Pro synthesizer. In Table 1, an over-
view on the automated syntheses, their conversion and
yield is given. The crude reaction mixture was purified
via semi-preparative radio-HPLC using isocratic condi-
tions (0.1 M ammonium acetate and acetonitrile (60/40;
v/v)) at a flow rate of 5 mL/min. An exemplary semi-
preparative HPLC chromatogram is outlined in Fig. 4a.
The precursor HAPTHI was found to be eluted at a re-
tention time of 4.5 min (k= 0.55) and the product
[
11
C]Me@HAPTHI at 7.6 min (k= 1.62), respectively.
Overall, seven large-scale radiosyntheses were per-
formed, yielding 2.2 ± 2.0 GBq (18.9 ± 13.3 %, corrected
Fig. 5 Dependence of the radiochemical yield of [
11
C]Me@HAPTHI (n3) on the
11
C-methylation agent a[
11
C]methyliodide or b
[
11
C]methyltriflate) in DMSO and 2-butanone using different bases (NaOH, triethylamine or TBAH) at 2-min reaction time
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 7 of 12
for decay to EOB) of sterile, formulated [
11
C]Me@HAPTHI
within 41 min including 5 min of radiopharmaceutical
quality control. A mean specific activity of 46.8 ±
28.5 GBq/μmol was found in the large-scale syntheses
(calculated using an HPLC-based method). A represen-
tative analytical HPLC chromatogram of the purified,
sterile [
11
C]Me@HAPTHI is shown in Fig. 4b. The reten-
tion times in the analytical HPLC assay were 3.37 min (k=
2.17) for precursor HAPTHI, 1.8 min (k=0.7)for
[
11
C]MeOH, 2.7 min (k= 1.55) for [
11
C]CH
3
OTf and
3.1 min (k=1.9)for[
11
C]CH
3
I, respectively. The product
[
11
C]Me@HAPTHI was eluted at a retention time of
4.38 min (k= 3.08). Radiochemical purity always exceeded
98 %. Osmolality and pH values were at all times found to
be in a physiological range. Residual solvent analysis using
GC revealed MEK <5 ppm and ACN <20 ppm, besides
8.5 % ethanol present in the product formulation (total
product volume 17.5 mL). Moreover, sterility and absence
of endotoxins was approved for all produced batches of
[
11
C]Me@HAPTHI upon complete decay of radioactivity
as in-process control.
Affinity and selectivity testings
Affinity of reference compounds (Me@HAPTHI and its
radiolabeling progenitor HAPTHI) was determined
using human NET membranes as K
d
= 0.21 ± 0.07 nM
for Me@HAPTHI and 24.2 ± 10.9 nM for HAPTHI,
respectively (n9 triplicates). For determination of
selectivity, the affinity of both reference substances was
assessed on human DAT and SERT membranes and re-
vealed >10 μM for both compounds for DAT and 409 ±
43 nM (Me@HAPTHI) and 10,274 ± 1207 nM (HAPTHI)
towards SERT, respectively, (n5 triplicates). Hence, se-
lectivity of Me@HAPTHI towards NET was determined
as DAT/NET >1947.6 and SERT/NET = 9757. Both values
clearly elucidate the ideal binding properties of our novel
NET PET ligand [
11
C]Me@HAPTHI.
LogD analysis, IAM chromatography and bloodbrain
barrier penetration
The lipophilicity of the novel NET PET radioligand
Me@HAPTHI was found to be in a decent range (logD =
2.27 ± 0.01) for a potential penetration of the BBB. The
precursor HAPTHI showed a logD value of 2.30 ± 0.01.
Additionally, BBB penetration experiments using IAM
chromatography revealed a permeability of P
m
=1.1
0.25 for Me@HAPTHI and P
m
= 1.14 ± 0.27 for the pre-
cursor HAPTHI, respectively. Both values were within the
identical, ideal range (P
m
=0.014.21) from other PET
tracers, known to easily penetrate the BBB [34].
Metabolic stability testing
Stability testing using human liver microsomes (n=4)re-
vealed no significant metabolism of [
11
C]Me@HAPTHI
within the tested timeframe. After 60 min, 99.6 ± 0.3 % of
the tracer was found to be still intact. Incubation of
[
11
C]Me@HAPTHI with pooled male rat liver micro-
somes revealed a higher metabolic degradation. The per-
centage of intact tracer over time is presented in Fig. 6.
Overall, 29.3 ± 1.9 % tracer was still intact after 1-h incu-
bation time. Thus, the stability of the novel NET PET
tracer [
11
C]Me@HAPTHI is encouraging in a human and
rodent setting and superior to the established reboxetine-
derived PET tracer [
18
F]FMeNER-D2.
Plasma protein binding
The mean percentage of plasma free fraction (ff ) and per-
centage of unspecific binding to the filter matrix of the
Centrifuge vials was determined. A plasma free fraction of
ff=8.2±0.3%(n= 7 triplicates) as well as an unspecific
filter retention of 51.26 ± 0.78 % was found. Overall, the ff
of our novel NET PET tracer [
11
C]Me@HAPTHI was in
the same range as that of [
11
C]ADAM [35].
In vitro autoradiography, immunohistochemistry and
Nissl staining
In the autoradiographic experiments, the highest uptake
of [
11
C]Me@HAPTHI was observed in NET-rich re-
gions identified with immunohistochemistry (Fig. 7).
Blocking was performed with non-radioactive NET li-
gands FMeNER-D2 and Me@HAPTHI in two different
concentrations each (100 nM, 1 μM). A concentration-
dependent binding displacement was observed using
human tissue samples for both cold competitors. In
Table 2, an overview on the percentage of specific dis-
placeable binding of [
11
C]Me@HAPTHI and fmol/mm
2
values of relative transporter protein density on the dif-
ferent tissue sections is given. All values are given in %
as mean n3 triplicates. Autoradiography of human
Table 1 Overview on the fully automated, large-scale
radiosyntheses of [
11
C]Me@HAPTHI
[
11
C]Me@HAPTHI (n= 7) Mean SD Median
Starting activity [
11
C]CO2 53.4 2.4 53.9
Trapped [
11
C]CH4 34.6 4.6 32
Trapped [
11
C]CH3I 29.6 2.4 29
Trapped [
11
C]CH3OTf in reactor 16.6 5.5 17.2
After quenching 8.8 3.6 8.9
Loss during injection in loop waste 1.0 0.5 0.8
Product [
11
C]Me@HAPTHI (EOS) 2.2 2.0 1.9
Yield (decay corr. to EOB) 13.7 13.5 15.9
Specific activity [GBq/μmol] (EOS) 43.4 29.7 59.2
Reaction conditions: [
11
C]MeOTf, NaOH, MEK, 2 mg/mL
precursor concentration
EOS end of synthesis, EOB end of bombardment
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 8 of 12
cerebellum revealed NET specific uptake in NET-rich
regions identified with IHC, though blocking experi-
ments were not possible due to the vast inhomogeneity
of the tissue samples. In human nucleus caudatus, a re-
gion known to be low in NET density, only unspecific
binding was observed.
Immunohistochemical staining was used to allocate
areas with high uptake in autoradiography with regions
known high NET abundance. Hence, the NET antibody-
dye complexes were found highly abundant in the heart
fibres, hippocampus, thalamus and hypothalamus and to
a minor extent in all other brain regions (Fig. 7). NET
specificity of staining was validated using a rabbit anti-
body isotype control.
Moreover, harvesting experiments with [
11
C]Me@HAP
THI using hNET expressing membranes were performed
according to the affinity testing protocol. Thereby, a
concentration-dependent displacement of [
11
C]Me@HA
PTHI was observed for all tested competitor substances
(cold FMeNER-D2 or Me@HAPTHI), and the counts
were corrected for decay (Fig. 8). Using Graph Pad
Prism, data correlation revealed akin-binding displacement
behaviour for both cold Me@HAPTHI as well as the
established NET ligand FMeNER-D2 (n3 triplicates).
Fig. 6 Metabolic stability of [
11
C]Me@HAPTHI against human and rat liver microsomes
Fig. 7 aNET-autoradiography and bimmunohistochemistry of [
11
C]Me@HAPTHI on 10 μm slices of human cortex, thalamus, hypothalamus,
cerebellum and nucleus caudatus as well as rat heart tissue and blocking with 100 nM FMeNER-D2, 1 μM FMeNER-D2, 100 nM Me@HAPTHI
and 1 μM Me@HAPTHI. The scale shows the radioactivity from high (red ) to low levels of radiotracer present on the Phosphor imager film
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 9 of 12
Discussion
[
11
C]Me@HAPTHI presents a large stride towards an
improved, novel, conveniently producible PET tracer for
NET imaging. This study comprises the first radiochemical
preparation, quality control and in vitro evaluation of this
novel candidate PET-tracer. We describe its affinity,
selectivity, lipophilicity and its potential to penetrate
the BBB as well as metabolic stability. Moreover, the
in vitro binding behaviour of [
11
C]Me@HAPTHI to
human NET cell membranes as well as human and ro-
dent tissue slices was examined.
The excellent affinity of Me@HAPTHI (K
d
hNET = 0.21 ±
0.07 nM) and exceptional selectivity of our candidate NET
PET ligand present the ideal ground for a further evalu-
ation of this tracer. Moreover, a lower non-specific bind-
ing can be expected, as the described radioligand is less
lipophilic than previous NET PET tracers based on rebox-
etine (logD Me@HAPTHI = 2.21, logD FMeNER-D2 =
2.73). Based on the in vitro data acquired, successful BBB
penetration by [
11
C]Me@HAPTHI may be expected. This
assumption is supported by immobilized artificial mem-
brane chromatography results showing Me@HAPTHI to
be within the discussed range of permeability P
m
values.
Additionally, the high radiochemical yields and feasible
radiosynthetic availability favour our newly developed
NET radioligand. The employed
11
C-methylation reac-
tion can be implemented at any PET facility with a
cyclotron. Hence, this study presents a large stride towards
a highly affine, selective and routinely available radiotracer.
Moreover, in vitro stability of [
11
C]Me@HAPTHI against
human liver microsomes, containing all human liver cyto-
chrome P450 enzymes, is excellent (99.6 ± 0.3 % intact
tracer after 60 min). In contrast, other existing PET tracers
show significant metabolic degradation within this time-
frame (e.g. [
11
C]MeNER, [
11
C]DASB or [
11
C]WAY-100635
[15, 44, 45]). Also in the rodent setting, where highly
increased turnover rates of the enzymes are present, a
Table 2 Overview of specific NET binding of the radioligand
[
11
C]Me@HAPTHI vs. Me@HAPTHI and FMeNER-D2 on rat and
human tissue origin
n3[
11
C]Me@HAPTHI
% BL-competitor fmol
Rat heart
FMeNER 1 μM 88.8 ± 11.2 <0.01
FMeNER 100nM 99.00 ± 0.07 <0.01
Me@HAPTHI 1 μM 92.5 ± 7.5* <0.01
Me@HAPTHI 100nM 104.5 ± 4.5 <0.01
Human cortex
FMeNER 1 μM 71.9 ± 7.9* 0.86
FMeNER 100nM 86.3 ± 11.2* <0.01
Me@HAPTHI 1 μM 66.3 ± 5.9* 1.32
Me@HAPTHI 100nM 82.1 ± 13.9* 0.02
Human thalamus
FMeNER 1 μM 68.36 ± 2.11 0.71
FMeNER 100nM 77.6 ± 9.8 0.47
Me@HAPTHI 1 μM 85.9 ± 18.5 0.09
Me@HAPTHI 100nM 92.5 ± 17.3 0.26
Human hypothalamus
FMeNER 1 μM 77.4 ± 14.5 0.02
FMeNER 100 nM 97.8 ± 14.6 0.11
Me@HAPTHI 1 μM 62.0 ± 3.6* 0.04
Me@HAPTHI 100 nM 83.7 ± 1.7* 0.05
Human hippocampus
FMeNER 1 μM 67.3 ± 8.2 <0.01
FMeNER 100 nM 97.1 ± 10.3 <0.01
Me@HAPTHI 1 μM 68.3 ± 5.3 <0.01
Me@HAPTHI 100 nM 84.1 ± 9.3 <0.01
Human nucleus caudatus
FMeNER 1 μM 107.6 ± 17.7 n.d.
FMeNER 100nM 102.6 ± 14.5 n.d.
Me@HAPTHI 1 μM 110.0 ± 21.0 n.d.
Me@HAPTHI 100nM 93.5 ± 12.5 nd
Human cerebellum
FMeNER 1 μM 108.2 ± 17.3 n.d.
FMeNER 100nM 103.9 ± 12.2 n.d.
Me@HAPTHI 1 μM 107.2 ± 20.8 n.d.
Me@HAPTHI 100nM 124.7 ± 10.8 n.d.
fmol values reflect calculated relative concentration (fmol/mm
2
) of transporter
protein). Limit of detection = 0.01 fmol; BL=baseline
n.d. not determined
*p< 0.05
Fig. 8 NET-binding of [
11
C]Me@HAPTHI on human NET expressing
cell membranes using a harvesting protocol. Competition was done
using different concentrations of Me@HAPTHI and FMeNER-D2 (1, 3,
10, 30, 100 and 1000 nM)
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 10 of 12
sufficient metabolic stability of [
11
C]Me@HAPTHI was ob-
served (29.26 ± 1.95 % intact, 60 min).
Furthermore, a plasma free fraction of 8.4 % was de-
termined in ultrafiltration experiments, which was in a
similar range with other clinically successful PET-tracers
(e.g. [
11
C]ADAM).
In vitro binding studies revealed specific displaceable
binding in human brain regions and rat heart, indicating
towards a promising further use of this tracer in in vivo
studies. Binding displacement was observed in competi-
tion experiments with different NET ligands FMeNER-D2
and Me@HAPTHI in a concentration-dependent manner.
The high radiotracer uptake areas matched with the high
NET-density regions identified by immunohistochemistry.
Therefore, specific NET uptake of [
11
C]Me@HAPTHI can
be affirmed. While this specific NET binding may be valid
on ex vivo tissue, the question of binding behaviour on a
cellular level was raised. Therefore, in vitro binding studies
on human NET membranes were performed in a cell
harvesting protocol. In these cell-based experiments,
which used the same parameters as autoradiography
studies (i.e. incubation time and buffer), a comparable
concentration-dependent binding displacement was found
for both competitors FMeNER-D2 and Me@HAPTHI.
Therefore, selective NET-uptake for our novel PET ligand
[
11
C]Me@HAPTHI could be confirmed on a cellular and
on a human and rat tissue level.
Thus, [
11
C]Me@HAPTHI was showing highly promis-
ing results in vitro so far and might therefore become an
improved, routine NET PET tracer. As a next step, small
animal experiments will be performed to further eluci-
date the in vivo behaviour of [
11
C]Me@HAPTHI.
Conclusions
A number of key properties have been discussed in the
presented study, indicating that the benzothiadiazole diox-
ide [
11
C]Me@HAPTHI presents a viable and improved
NET PET tracer.
We demonstrated its outstanding affinity and selectivity,
its great stability in human liver microsomes, as well as
promising results from in vitro autoradiography. There-
fore, these data encourage us for an in vivo application of
this compound in small animal PET experiments in the
future. On these grounds, [
11
C]Me@HAPTHI might im-
prove clinical NET imaging.
Additional file
Additional file 1: Supplementary data on affinity testing, metabolic
stability assessments and autoradiography. Detailed methods for
synthesis of precursor and reference compounds, the affinity testing of
the new radiolabelled ligand via NET-expressing membrane binding
protocol, as well as detailed procedures to autoradiography,
immunohistochemistry and metabolic stability testings.
Competing interests
Dr. Alexander Hoepping is a full employee at the ABX Advanced Biochemical
Compounds. All other authors declare that they have no competing interests.
Authorscontributions
CRM performed all radiosyntheses and preclinical in vitro experiments,
autoradiography, immunohistochemistry and writing of the paper. NB
contributed to all radiosyntheses and metabolite studies. CP contributed to
in vitro autoradiography and immunohistochemistry. SF contributed to the
affinity and selectivity testing procedures. CV contributed to IAM
chromatography experiments and plasma free fraction. AH performed
the synthesis of the cold reference standard Me@HAPTHI and the
precursor HAPTHI. RL participated in the design of the study and
proofread the manuscript. MH designed parts of the research and
proofread the manuscript. MM conceived and supervised the preclinical
experiments and proofread the manuscript. WW conceived and
supervised the radiosyntheses and proofread the manuscript. All authors
read and approved the manuscript.
Acknowledgements
The authors would like to thank Vanessa Fröhlich and Thomas Zenz for their
practical and technical support. The authors are grateful to Marie Spies, MD,
for native English editing.
Author details
1
Department of Biomedical Imaging and Image-guided Therapy, Division of
Nuclear Medicine, Medical University of Vienna, Vienna, Austria.
2
Department
of Inorganic Chemistry, University of Vienna, Vienna, Austria.
3
Faculty of Life
Sciences, Department of Technology and Biopharmaceutics, University of
Vienna, Vienna, Austria.
4
ABX Advanced Biochemical Compounds, Radeberg,
Germany.
5
Department of Psychiatry and Psychotherapy, Division of
Biological Psychiatry, Medical University of Vienna, Vienna, Austria.
Received: 10 April 2015 Accepted: 22 May 2015
References
1. Zhou J. Norepinephrine transporter inhibitors and their therapeutic
potential. Drugs Future. 2004;29:123544.
2. Zhou J, Zhang A, Klaess T, Johnson KM, Wang CZ, Ye YP, et al. Biaryl
analogues of conformationally constrained tricyclic tropanes as potent and
selective norepinephrine reuptake inhibitors: synthesis and evaluation of
their uptake inhibition at monoamine transporter sites. J Med Chem.
2003;46:19972007.
3. Curatolo P, DAgati E, Moavero R. The neurobiological basis of ADHD. Ital J
Pediatr. 2010;36:79.
4. Mash DC, Ouyang Q, Qin Y, Pablo J. Norepinephrine transporter
immunoblotting and radioligand binding in cocaine abusers. J Neurosci
Methods. 2005;143:7985.
5. Schlessinger A, Geier E, Fan H, Irwin JJ, Shoichet BK, Giacomini KM, et al.
Structure-based discovery of prescription drugs that interact with the
norepinephrine transporter, NET. Proc Natl Acad Sci U S A. 2011;108:158105.
6. Vazey EM, Aston-Jones G. The emerging role of norepinephrine in cognitive
dysfunctions of Parkinsons disease. Front Behav Neurosci. 2012;6:48.
7. Moldovanova I, Schroeder C, Jacob G, Hiemke C, Diedrich A, Luft FC, et al.
Hormonal influences on cardiovascular norepinephrine transporter
responses in healthy women. Hypertension. 2008;51:12039.
8. Harik S, Duckrow R, LaManna J, Rosenthal M, Sharma V, Banerjee S. Cerebral
compensation for chronic noradrenergic denervation induced by locus
ceruleus lesion: recovery of receptor binding, isoproterenol- induced adenylate
cyclase activity, and oxidative metabolism. J Neurosci. 1981;1:6419.
9. Rommelfanger KS, Edwards GL, Freeman KG, Liles LC, Miller GW,
Weinshenker D. Norepinephrine loss produces more profound motor
deficits than MPTP treatment in mice. Proc Natl Acad Sci U S A.
2007;104:138049.
10. Wilson AA, Patrick Johnson D, Mozley D, Hussey D, Ginovart N, Nobrega J,
et al. Synthesis and in vivo evaluation of novel radiotracers for the in vivo
imaging of the norepinephrine transporter. Nucl Med Biol. 2003;30:8592.
11. Takano A, Gulyás B, Varrone A, Halldin C. Comparative evaluations of
norepinephrine transporter radioligands with reference tissue models in
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 11 of 12
rhesus monkeys: (S, S)-[18F]FMeNER-D2 and (S, S)-[11C]MeNER. Eur J Nucl
Med Mol Imaging. 2009;36:188591.
12. Schou M, Halldin C, Sóvágó J, Pike VW, Hall H, Gulyás B, et al. PET evaluation
of novel radiofluorinated reboxetine analogs as norepinephrine transporter
probes in the monkey brain. Synapse. 2004;53:5767.
13. Gulyás B, Brockschnieder D, Nag S, Pavlova E, Kása P, Beliczai Z, et al. The
norepinephrine transporter (NET) radioligand (S, S)-[18F]FMeNER-D2 shows
significant decreases in NET density in the human brain in Alzheimers
disease: a post-mortem autoradiographic study. Neurochem Int.
2010;56:78998.
14. Rami-Mark C, Zhang MR, Mitterhauser M, Lanzenberger R, Hacker M, Wadsak
W. [
18
F]FMeNER-D2: reliable fully-automated synthesis for visualization of
the norepinephrine transporter. Nucl Med Biol. 2013;40:104954.
15. Schou M, Zoghbi S, Shetty H, Shchukin E, Liow J-S, Hong J, et al. Investigation
of the metabolites of (S, S)-[11C]MeNER in humans, monkeys and rats. Mol
Imaging Biol. 2009;11:2330.
16. Zeng F, Jarkas N, Stehouwer JS, Voll RJ, Owens MJ, Kilts CD, et al. Synthesis,
in vitro characterization, and radiolabeling of reboxetine analogs as
potential PET radioligands for imaging the norepinephrine transporter.
Bioorg Med Chem. 2008;16:78393.
17. Zeng F, Mun J, Jarkas N, Stehouwer JS, Voll RJ, Tamagnan GD, et al.
Synthesis, radiosynthesis, and biological evaluation of carbon-11 and
fluorine-18 labeled reboxetine analogues: potential positron emission
tomography radioligands for in vivo imaging of the norepinephrine
transporter. J Med Chem. 2008;52:6273.
18. Zeng F, Stehouwer JS, Jarkas N, Voll RJ, Williams L, Camp VM, et al.
Synthesis and biological evaluation of 2β,3α-(substituted
phenyl)nortropanes as potential norepinephrine transporter imaging agents.
Bioorg Med Chem Lett. 2007;17:30447.
19. Mark C, Bornatowicz B, Mitterhauser M, Hendl M, Nics L, Haeusler D, et al.
Development and automation of a novel NET-PET tracer: [
11
C]Me@APPI.
Nucl Med Biol. 2013;40:295303. doi:10.1016/j.nucmedbio.2012.11.009.
20. ONeill DJ, Adedoyin A, Bray JA, Deecher DC, Fensome A, Goldberg JA, et al.
Discovery of novel selective norepinephrine inhibitors: 1-(2-morpholin-2-
ylethyl)-3-aryl-1,3-dihydro-2,1,3-benzothiadiazole 2,2-dioxides (WYE-114152).
J Med Chem. 2011;54:682431. doi:10.1021/jm200733r.
21. O'Neill DJ, Adedoyin A, Alfinito PD, Bray JA, Cosmi S, Deecher DC, et al.
Discovery of novel selective norepinephrine reuptake inhibitors: 4-[3-aryl-
2,2-dioxido-2,1,3-benzothiadiazol-1(3H)-yl]-1-(methylamino)butan-2-ols
(WYE-103231). J Med Chem. 2010;53:451121.
22. Larsen P, Ulin J, Dahlstrøm K, Jensen M. Synthesis of [
11
C]iodomethane by
iodination of [
11
C]methane. Appl Radiat Isot. 1997;48:1537.
23. Kniess T, Rode K, Wuest F. Practical experiences with the synthesis of
[
11
C]CH3I through gas phase iodination reaction using a TRACERlabFXC
synthesis module. Appl Radiat Isot. 2008;66:4828.
24. Jewett DM. A simple synthesis of [
11
C]methyl triflate. Int J Radiat Appl
Instrum Part A. 1992;43:13835.
25. Council of Europe. Radioactive pharmaceuticals (European Pharmacopoeia
(Europäisches Arzneibuch). 6th ed. Vienna: Verlag Österreich GmbH; 2008.
26. Zeng F, Jarkas N, Owens MJ, Kilts CD, Nemeroff CB, Goodman MM.
Synthesis and monoamine transporter affinity of front bridged tricyclic
3β-(4-halo or 4-methyl)phenyltropanes bearing methylene or
carbomethoxymethylene on the bridge to the 2β-position. Bioorg
Med Chem Lett. 2006;16:46613.
27. Tejani-Butt SM. [3H]nisoxetine: a radioligand for quantitation of
norepinephrine uptake sites by autoradiography or by homogenate
binding. J Pharmacol Exp Ther. 1992;260:42736.
28. Donovan SF, Pescatore MC. Method for measuring the logarithm of the
octanol-water partition coefficient by using short octadecyl-poly(vinyl
alcohol) high-performance liquid chromatography columns. J Chromatogr
A. 2002;952:4761.
29. Naik P, Cucullo L. In vitro bloodbrain barrier models: current and
perspective technologies. J Pharm Sci. 2012;101:133754.
30. Neuhaus W, Freidl M, Szkokan P, Berger M, Wirth M, Winkler J, et al. Effects
of NMDA receptor modulators on a bloodbrain barrier in vitro model.
Brain Res. 2011;1394:4961.
31. Yoon CH, Kim SJ, Shin BS, Lee KC, Yoo SD. Rapid screening of bloodbrain
barrier penetration of drugs using the immobilized artificial membrane
phosphatidylcholine column chromatography. J Biomol Screen.
2006;11:1320.
32. Tavares AA, Lewsey J, Dewar D, Pimlott SL. Radiotracer properties
determined by high performance liquid chromatography: a potential tool
for brain radiotracer discovery. Nucl Med Biol. 2012;39:12735.
33. Rami-Mark C, Bornatowicz B, Fink C, Otter P, Ungersboeck J, Vraka C, et al.
Synthesis, radiosynthesis and first in vitro evaluation of novel PET-tracers for
the dopamine transporter: [(11)C]IPCIT and [(18)F]FE@IPCIT. Bioorg Med
Chem Lett. 2013;21:75629.
34. Vraka C, Nics L, Weiss V, Wagner K-H, Hacker M, Wadsak W, et al. Combination
of high throughput HPLC methods for rapid prediction of blood brain barrier
penetration of newly developed radiotracersEANM 14. Eur J Nucl Med Mol
Imaging. 2014;41:442.
35. Huang Y, Hwang D-R, Narendran R, Sudo Y, Chatterjee R, Bae S-A, et al.
Comparative evaluation in nonhuman primates of five PET radiotracers for
imaging the serotonin transporters[
11
C]McN 5652, [
11
C]ADAM, [
11
C]DASB,
[
11
C]DAPA, and [
11
C]AFM. J Cereb Blood Flow Metab. 2002;22:137798.
36. Gandelman MS, Baldwin RM, Zoghbi SS, Zea-Ponce Y, Innis RB. Evaluation of
ultrafiltration for the free-fraction determination of single photon emission
computed tomography (SPECT) radiotracers: beta-CIT, IBF, and iomazenil.
Thai J Pharm Sci. 1994;83:10149.
37. Price JC, Mayberg HS, Dannals RF, Wilson AA, Ravert HT, Sadzot B.
Measurement of benzodiazepine receptor number and affinity in humans
using tracer kinetic modeling, positron emission tomography, and
[
11
C]flumazenil. Journal of cerebral blood flow and Metabolism.
1993;13:65667.
38. Sadzot B, Price JC, Mayberg HS, Douglass KH, Dannals RF, Lever JR, et al.
Quantification of human opiate receptor concentration and affinity using
high and Low specific activity [
11
C]diprenorphine and positron emission
tomography. J Cereb Blood Flow Metab. 1991;11:20419.
39. Schou M, Halldin C, Pike VW, Mozley PD, Dobson D, Innis RB, et al.
Post-mortem human brain autoradiography of the norepinephrine
transporter using (S, S)-[18F]FMeNER-D2. Eur Neuropsychopharmacol.
2005;15:51720.
40. Philippe C, Haeusler D, Fuchshuber F, Spreitzer H, Viernstein H, Hacker M,
et al. Comparative autoradiographic in vitro investigation of melanin
concentrating hormone receptor 1 ligands in the central nervous system.
Eur J Pharmacol. 2014;735:17783.
41. Kádár A, Wittmann G, Liposits Z, Fekete C. Improved method for
combination of immunocytochemistry and Nissl staining. J Neurosci
Methods. 2009;184:1158.
42. Fukuda T, Koelle GB. The cytological localization of intracellular neuronal
acetylcholinesterase. J Biophys Biochem Cytol. 1959;5:43340.
43. Immunohistochemistry HF. Current protocols in immunology. 2001.
44. Någren K, Halldin C, Pike VW, Allonen T, Hietala J, Swahn CG, et al. Radioactive
metabolites of the 5-HT1A receptor pet radioligand, [carbonyl-11C]way-100635,
measured in human plasma samples. J Label Compd Radiopharm.
2001;44:S4724.
45. Parsey RV, Ojha A, Ogden RT, Erlandsson K, Kumar D, Landgrebe M, et al.
Metabolite considerations in the in vivo quantification of serotonin
transporters using 11C-DASB and PET in humans. J Nucl Med.
2006;47:1796802.
Submit your manuscript to a
journal and benefi t from:
7 Convenient online submission
7 Rigorous peer review
7 Immediate publication on acceptance
7 Open access: articles freely available online
7 High visibility within the fi eld
7 Retaining the copyright to your article
Submit your next manuscript at 7 springeropen.com
Rami-Mark et al. EJNMMI Research (2015) 5:34 Page 12 of 12
... The IAM (Immobilized Artificial Membrane) chromatography was slightly modified from previously published data [23][24][25]. The retention times of the analytes (reference tracer) on the IAM.PC.DD2 stationary phase (150 × 4.6 mm, column-batch: 45873 and 49161, REGIS Technologies, USA) were measured by HPLC with an isocratic flow rate of 1 ml/min and wavelengths of 254 nm and 285 m using four runs with different eluent ratios (50/50, 55/45, 60/40 and 65/ 35) of 0.01 M sodium phosphate buffer, pH 7.0 (sodium phosphate dibasic dehydrate CAS 10028-24-7 and sodium phosphate monobasic monohydrate (CAS10049-21-5), all from Sigma-Aldrich, St Louis, USA, water from Milli-Q® Integral Water Purification System (Merck Millipore, Billerica, USA) and acetonitrile (LiChrosol®Reag. ...
... K IAM and Pm have been used together with the logP to predict BBB penetration in recent publications [23][24][25][39][40][41]. However, the emphasis was only the permeability value, but additional comprehensive reference values were missing. ...
Article
Introduction: Due to the high candidate exclusion rate during a drug development process, an early prediction of the pharmacokinetic behavior would be needed. Accordingly, high performance bioaffinity chromatography (HPBAC) approaches are growing in popularity, however, there is a lack of knowledge and no consensus about the relation between HPBAC measurements, in vivo distribution and blood brain barrier (BBB) penetration behavior. With respect to radiotracers, there is almost no reference data available for plasma protein binding (PPB), permeability (Pm) and the membrane coefficient (KIAM). Thus, this study was aimed at exploring the relevance of measuring PPB, Pm and KIAM for the prediction of BBB penetration. Methods: Measurements of %PPB, Pm and KIAM were performed using HPBAC. In total, 113 compounds were tested, 43 with brain uptake, 30 not showing brain uptake and 40 with known interactions with efflux transporters. Additionally, ClogP and HPLC logPowpH7.4 data were collected. Results: %PPB, KIAM, Pm and ClogP values were in the same range for each of the three groups. A significant difference was observed for the HPLC logPowpH7.4 between CNS penetrating drug group (CNSpos) and the non-penetrating drug group (CNSneg), as well as for the CNSneg towards the drug group interacting with efflux transporters (DRUGefflux). However, as the other experimental data, also the HPLC logPowpH7.4 showed a broad overlapping of the single values between the groupings. Conclusion: Experimental reference values (logP, Pm, KIAM & PPB) of commonly used PET tracers and drugs showing different BBB penetration behavior are provided. The influence of the logP on brain uptake depends strongly on the selected method. However, using a single parameter (experimental or calculated) to predict BBB penetration or for the classification of drug groups is inexpedient.
... Researchers concluded that 11 Clabeled FAPI-01 and 11 C-FAPI-02 could be used in the clinic because they increased tumor uptake and retained tumors longer than conventional FAPIs, suggesting that 11 C-labeled FAPI treatment could be used in the clinic and benefit from its physical imaging and short half-life. Interestingly, 11 C-labeled tracers can pass through the blood-brain barrier more easily and image the brain glioma more quickly than [ 68 Ga] Ga-DOTA-FAPI-04 in U87MG tumor xenografts [112]. Additionally, an optimized linker was developed to create 64 Cu-labeled compounds targeting FAP and PSMA, potentially allowing them to treat and image various cancers. ...
Article
Cancer cells are surrounded by a complex and highly dynamic tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs), a critical component of TME, contribute to cancer cell proliferation as well as metastatic spread. CAFs express a variety of biomarkers, which can be targeted for detection and therapy. Most importantly, CAFs express high levels of fibroblast activation protein (FAP) which contributes to progression of cancer, invasion, metastasis, migration, immunosuppression, and drug resistance. As a consequence, FAP is an attractive theranostic target. In this review, we discuss the latest advancement in targeting FAP in oncology using theranostic biomarkers and imaging modalities such as single-photon emission computed tomography (SPECT), positron emission tomography (PET), computed tomography (CT), fluorescence imaging, and magnetic resonance imaging (MRI).
... Furthermore, the tracer shows excellent metabolic stability in human liver microsomes with > 99% intact after 60 min incubation. Autoradiography on human brain tissue has revealed NET-specific uptake in NETrich regions (Rami-Mark et al. 2015). Notably, in addition to CNS uptake, significant cardiac uptake of 11 C-Me@ HAPTHI has been observed, albeit with a more lipophilic Fig. 3 a Autoradiography of left ventricular short axis slices from rats following administration of 18 F-AF78 with (right) and without (left) NET blockade. ...
Article
Full-text available
The norepinephrine transporter (NET) is a major target for the evaluation of the cardiac sympathetic nerve system in patients with heart failure and Parkinson's disease. It is also used in the therapeutic applications against certain types of neuroendocrine tumors, as exemplified by the clinically used 123/131I-MIBG as theranostic single-photon emission computed tomography (SPECT) agent. With the development of more advanced positron emission tomography (PET) technology, more radiotracers targeting NET have been reported, with superior temporal and spatial resolutions, along with the possibility of functional and kinetic analysis. More recently, fluorine-18-labelled NET tracers have drawn increasing attentions from researchers, due to their longer radiological half-life relative to carbon-11 (110 min vs. 20 min), reduced dependence on on-site cyclotrons, and flexibility in the design of novel tracer structures. In the heart, certain NET tracers provide integral diagnostic information on sympathetic innervation and the nerve status. In the central nervous system, such radiotracers can reveal NET distribution and density in pathological conditions. Most radiotracers targeting cardiac NET-function for the cardiac application consistent of derivatives of either norepinephrine or MIBG with its benzylguanidine core structure, e.g. 11C-HED and 18F-LMI1195. In contrast, all NET tracers used in central nervous system applications are derived from clinically used antidepressants. Lastly, possible applications of NET as selective tracers over organic cation transporters (OCTs) in the kidneys and other organs controlled by sympathetic nervous system will also be discussed.
... The availability of PET markers for quantifying NET fiber density in the human brain means LC fiber density could be assessed in imaging studies [53][54][55][56]. An agingrelated decline in LC NET [54] consistent with LC volume loss with age in humans [4] has been observed. ...
Article
Full-text available
Background: The earliest brain pathology related to Alzheimer's disease (AD) is hyperphosphorylated soluble tau in the noradrenergic locus coeruleus (LC) neurons. Braak characterizes five pretangle tau stages preceding AD tangles. Pretangles begin in young humans and persist in the LC while spreading from there to other neuromodulatory neurons and, later, to the cortex. While LC pretangles appear in all by age 40, they do not necessarily result in AD prior to death. However, with age and pretangle spread, more individuals progress to AD stages. LC neurons are lost late, at Braak stages III-IV, when memory deficits appear. It is not clear if LC hyperphosphorylated tau generates the pathology and cognitive changes associated with preclinical AD. We use a rat model expressing pseudohyperphosphorylated human tau in LC to investigate the hypothesis that LC pretangles generate preclinical Alzheimer pathology. Methods: We infused an adeno-associated viral vector carrying a human tau gene pseudophosphorylated at 14 sites common in LC pretangles into 2-3- or 14-16-month TH-Cre rats. We used odor discrimination to probe LC dysfunction, and we evaluated LC cell and fiber loss. Results: Abnormal human tau was expressed in LC and exhibited somatodendritic mislocalization. In rats infused at 2-3 months old, 4 months post-infusion abnormal LC tau had transferred to the serotonergic raphe neurons. After 7 months, difficult similar odor discrimination learning was impaired. Impairment was associated with reduced LC axonal density in the olfactory cortex and upregulated β1-adrenoceptors. LC infusions in 14-16-month-old rats resulted in more severe outcomes. By 5-6 months post-infusion, rats were impaired even in simple odor discrimination learning. LC neuron number was reduced. Human tau appeared in the microglia and cortical neurons. Conclusions: Our animal model suggests, for the first time, that Braak's hypothesis that human AD originates with pretangle stages is plausible. LC pretangle progression here generates both preclinical AD pathological changes and cognitive decline. The odor discrimination deficits are similar to human odor identification deficits seen with aging and preclinical AD. When initiated in aged rats, pretangle stages progress rapidly and cause LC cell loss. These age-related outcomes are associated with a severe learning impairment consistent with memory decline in Braak stages III-IV.
... Subsequently, [ 11 C]CO2 was converted into [ 11 C]CH4 by a Ni-catalyzed reduction with H2 at 400 °C. The [ 11 C]CH3I was produced using gas phase conversion, as described in a previous study [18]. In short, the resulting [ 11 C]CH4 was reacted with sublimated iodine at 738 °C in a recirculating process for 5 min to give [ 11 C]CH3I. ...
Article
Full-text available
The tracer [[11C]meta-Hydroxyephedrine ([[11C]mHED) is one of the most applied PET tracers for cardiac imaging, whose radiosynthesis was already reported in 1990. While not stated in the literature, separation difficulties and an adequate formulation of the product are well known challenges in its production. Furthermore, the precursor (metaraminol) is also a substrate for the norepinephrine transporter, and can therefore affect the image quality. This study aims at optimizing the synthetic process of [[11C]mHED and investigating the effect of the apparent molar activity (sum of mHED and metaraminol) in patients and animals. The main optimization was the improved separation through reverse phase-HPLC by a step gradient and subsequent retention of the product on a weakly-cationic ion exchange cartridge. The µPET/µCT was conducted in ten rats (ischemic model) and the apparent molar activity was correlated to the VOI- and SUV-ratio of the myocardium/intra-ventricular blood pool. Moreover, nine long-term heart transplanted and five Morbus Fabry patients underwent PET and MRI imaging for detection of changes in the sympathetic innervation. In summary, the fully-automated synthesis and optimized purification method of [[11C]mHED is easily applicable and reproducible. Moreover, it was shown that the administered apparent molar activities had a negligible effect on the imaging quality.
... In the case of DOPA, metaraminol and methionine no retention was observed. Based on the fact that three different logP values are available in the literature for the internal standards (toluene and triphenylene), results are presented in four columns: receptor ligands, NET ligands, IPCIT derivatives and MCHR1 antagonist were previously published or equivalent results were found using the same HPLC method[14][15][16][17][18]. Additionally, the calculated logP (Clog) values for each compound are given. ...
Article
Introduction: There is an increasing demand for high throughput methods at early stages of preclinical radioligand development, in order to predict pharmacokinetic properties (e.g., biodistribution) and blood brain barrier (BBB) penetration. One of the most important physicochemical properties is the lipophilicity, measured by means of shake-flask (logP) or HPLC methods. Yet, a plethora of experimental methods are described in the literature for the determination of logP values. These varying methods often lead to different results for one identical compound, which complicates any comparison or prediction for subsequent preclinical studies. However, a standardized and internationally applied and accepted database with logP values for a reliable comparison of the lipophilic character of radiotracers is still missing. Method: Lipophilicity measurements were performed with 121 molecules using a high throughput HPLC method and ClogP values were calculated using ChemBioDraw?. Furthermore, logP measurements for six representative radiotracers were performed with the conventional shake-flask method and the results were statistically compared to the ClogP and HPLC logP results. Different logP thresholds, suggesting optimal BBB penetration according to literature, were selected and put into relation with the acquired HPLC logP and ClogP values of cerebral tracers. Results: The results of the tested compounds ranged from -2.1 to 5.4 with the applied HPLC method. The acquired database comprises ClogP values of the whole set of compounds ranging from -4.11 to 6.12. LogP data from different methods were not comparable. The correlation of the obtained logP data to thresholds suggesting an optimal brain uptake resulted in a high number of false positive classifications. Conclusion: The logP determination for prediction of BBB penetration is obsolete. The extensive database, including clinical relevant radiotracers, can be used as comparative set of values for preclinical studies, and serves as a basis for further critical discussions concerning the eligibility of logP.
... However, if its pharmacokinetic profile turns out to be suitable, its subnanomolar affinity for NET may eventually lead to an improved specific-to-non-specific binding ratio in vivo. 236 Serotonin transporter (SERT) ( Table 4, Schemes 35-37) Alterations of the SERT have been implicated mainly in neuropsychiatric disorders such as depression, anxiety, and suicide. SERT is also the target of the SSRI antidepressant drugs. ...
Chapter
Positron emission tomography (PET) is a quantitative molecular imaging technology based on radiotracers typically labeled with ¹¹C and ¹⁸F that can quantify biochemical processes within the living human brain. Over the last decade, PET imaging has been increasingly used to support neuroscience drug discovery by providing direct measurement of the drug–target interaction in the brain. This article will provide an overview of the available PET tracers for clinical studies and of the recent advances in the identification of novel PET tracers, will discuss some of the challenges and limits to develop new PET tracers, and will eventually suggest future areas of interest.
... Appropriate kinetic modeling is also indispensable for analyzing kinetics around the sympathetic terminal. New radiotracers such as 11 C-Me@HAPTHI offer promise for better demonstration of the NET system, 4 and these preclinical investigations may lead to development of clinically effective parameters that reflect NET activity. Finally, electrocardiographic and respiratory gating may be required for analyzing a moving heart. ...
Article
Full-text available
Supported by their involvement in many neurodegenerative disorders, muscarinic acetylcholine receptors (mAChRs) are an interesting target for PET imaging. Nevertheless, no radiotracer is established in clinical routine. Within this work we aim to develop novel PET tracers based on the structure of arecoline. Fifteen novel arecoline derivatives were synthesized, characterized and tested for their affinity to the mAChRs M1-M5 and the conceivable off-target acetylcholinesterase. Five arecoline derivatives and arecoline were labeled with carbon-11 in good yields. Arecaidine diphenylmethyl ester (3b), arecaidine bis(4-fluorophenyl)methyl ester (3c) and arecaidine (4-bromophenyl)(4-fluorophenyl)methyl ester (3e) showed a tremendous gain in mAChR affinity compared to arecoline and a pronounced subtype selectivity for M1. Metabolic stability and serum protein binding of [¹¹C]3b and [¹¹C]3c were in line with properties of established brain tracers. Nonspecific binding of [¹¹C]3c was prevalent in kinetic and endpoint experiment on living cells as well as in autoradiography on native mouse brain sections, which motivates us to decrease the lipophilicity of this substance class prior to in vivo experiments.
Article
In drug development, biomarkers for cerebral applications have a lower success rate compared to cardiovascular drugs or tumor therapeutics. One reason is the missing blood brain barrier penetration, caused by the tracer's interaction with efflux transporters such as the P-gp (MDR1 or ABCB1). Aim of this study was the development of a reliable model to measure the interaction of radiotracers with the human efflux transporter P-gp in parallel to the radiolabeling process. LigandTracer® Technology was used with the wildtype cell line MDCKII and the equivalent cell line overexpressing human P-gp (MDCKII-hMDR1). The method was evaluated based on established PET tracers with known interaction with the human P-gp transporter and in nanomolar concentration (15 nM). [¹¹C]SNAP-7941 and [¹⁸F]FE@SNAP were used as P-gp substrates by comparing the real-time model with an uptake assay and μPET images. [¹¹C]DASB [¹¹C]Harmine, [¹⁸F]FMeNER,[¹⁸F]FE@SUPPY and [¹¹C]Me@HAPTHI were used as tracers without interactions with P-gp in vitro. However, [¹¹C]Me@HAPTHI shows a significant increase in SUV levels after blocking with Tariquidar. The developed real-time kinetic model uses directly PET tracers in a compound concentration, which is reflecting the in vivo situation. This method may be used at an early stage of radiopharmaceutical development to measure interactions to P-gp before conducting animal experiments.
Article
Full-text available
In neurodegenerative diseases and neuropsychiatric disorders dysregulation of the norepinephrine transporter (NET) has been reported. For visualization of NET availability and occupancy in the human brain PET imaging can be used. Therefore, selective NET-PET tracers with high affinity are required. Amongst these, [(18)F]FMeNER-D2 is showing the best results so far. Furthermore, a reliable fully automated radiosynthesis is a prerequisite for successful application of PET-tracers. The aim of this work was the automation of [(18)F]FMeNER-D2 radiolabelling for subsequent clinical use. The presented study comprises 25 automated large-scale syntheses, which were directly applied to healthy volunteers and adult patients suffering from attention deficit hyperactivity disorder (ADHD). Procedures: Synthesis of [(18)F]FMeNER-D2 was automated within a Nuclear Interface Module. Starting from 20-30GBq [(18)F]fluoride, azeotropic drying, reaction with Br2CD2, distillation of 1-bromo-2-[(18)F]fluoromethane-D2 ([(18)F]BFM) and reaction of the pure [(18)F]BFM with unprotected precursor NER were optimized and completely automated. HPLC purification and SPE procedure were completed, formulation and sterile filtration were achieved on-line and full quality control was performed. Purified product was obtained in a fully automated synthesis in clinical scale allowing maximum radiation safety and routine production under GMP-like manner. So far, more than 25 fully automated syntheses were successfully performed, yielding 1.0-2.5GBq of formulated [(18)F]FMeNER-D2 with specific activities between 430 and 1707GBq/μmol within 95min total preparation time. A first fully automated [(18)F]FMeNER-D2 synthesis was established, allowing routine production of this NET-PET tracer under maximum radiation safety and standardization.
Article
Full-text available
Parkinson's disease (PD) is the second most common neurodegenerative disorder, affecting 1% of the population over age 60. In those patients cognitive dysfunction is a persistent issue that impairs quality of life and productivity. Neuropathological studies demonstrate significant damage in brain regions outside the nigral dopamine (DA) system, including early degeneration of locus coeruleus norepinephrine (LC-NE) neurons, yet discussion of PD and treatment focus has remained dopaminergic-based. Motor symptoms benefit from DA replacement for many years, but other symptoms including several cognitive deficits continue unabated. Recent interest in non-DA substrates of PD highlights early involvement of LC-NE neurons and provides evidence for a prodromal phase, with cognitive disturbance, even in sporadic PD. We outline insights from basic research in LC-NE function to clinical and pathological evidence highlighting a role for NE in PD cognitive dysfunction. We propose that loss of LC-NE regulation, particularly in higher cortical regions, critically underlies certain cognitive dysfunctions in early PD. As a major unmet need for patients, research and use of NE drugs in PD may provide significant benefits for cognitive processing.
Article
Full-text available
[11C]iodomethane was synthesized by iodination of [11C]methane with iodine. The reaction was carried out in a system where [11C]methane, helium and iodine vapours were mixed and heated. The formed [11C]iodomethane was continuously removed from the reaction mixture and the unchanged [11C]methane was recirculated into the reaction chamber by a pump. The decay-corrected radiochemical yield was 83% after a production time of 7 min (from trapped [11C]methane). The specific radioactivity was better than 550 GBq/μmol (15 Ci/mmol) at the end of the synthesis.
Article
Measures of the percentage of radioactivity represented by parent radioligand in human plasma during PET experiments with [carbonyl-11C]WAY-100635 have been compared across 3 centres using 2 different methods. Test-retest reliability has been examined in 5 subjects in one centre. Good agreement is found for inter-centre and test-retest measures.
Article
IntroductionPresent data indicate that merging beneficial structural elements from previously published DAT-ligands highest DAT affinity, selectivity and a suitable metabolic profile should be achieved. This combination led to the development of IPCIT and FE@IPCIT.Methods Precursor synthesis was done starting from cocaine in a six step reaction. O-[11C]-methylation was established using [11C]methyl iodide, optimized and subsequently automated. Small scale 18F-fluroroethylation as well as optimization of reaction parameters and automation were performed. Affinity and selectivity of the candidate substances were tested in standard binding experiments on human membranes. Metabolic stability and blood–brain-barrier (BBB) penetration were determined.ResultsPrecursor compound, IPCITacid, and reference compounds, IPCIT and FE@IPCIT, were obtained in 4.9%, 12.7% and 4.1% yield, respectively. Automated radiosynthesis of [11C]IPCIT yielded 1.9 ± 0.7 GBq (12.5 ± 4%, corrected for decay). Optimum parameters for 18F-fluoroethylation were 110 °C for 15 min under TBAH catalysis, yielding 67 ± 16% radiochemical incorporation. Affinity was determined as 1.7 ± 0.6 nM for IPCIT, 1.3 ± 0.2 nM for FE@IPCIT and 37 ± 13 nM for the precursor molecule, IPCIT-acid. Results from in vitro and in silico evaluations revealed high stability but also high lipophilicity.Conclusion Present data indicate high affinity and stability of both IPCIT and FE@IPCIT. Radiolabelling, optimization of reaction parameters and automation succeeded. On the other hand, data concerning BBB-penetration are not promising.
Article
Introduction: The norepinephrine transporter (NET) is an important target for research in neurology and psychology and is involved in the pathophysiology of many neurodegenerative diseases such as Alzheimer's disease and attention deficient hyperactivity disorder. For visualization of NET abundance and deregulation, a novel PET tracer--[(11)C]Me@APPI--has been developed. Methods: For precursor synthesis, a 4-step synthesis starting from N-phenyl-o-phenylenediamine was set up. Radiosynthesis was established and optimized using standard methods and subsequently automated in a GE TRACERlabFx C Pro synthesizer. Preclinical testing was performed comprising affinity and selectivity testing on human membranes as well as stability and blood-brain-barrier-penetration using in-vitro models. Results: Precursor molecule (APPI:0) and reference compound (Me@APPI) were synthesized with 26.5% and 21.4% overall yield, respectively. So far, 1.25±0.72 GBq [(11)C]Me@APPI with 54.35±7.80 GBq/μmol specific activity were produced (n=11). Affinity of reference compounds was determined as 8.08±1.75 nM for Me@APPI and 19.31±2.91 nM for APPI:0, respectively (n≥9). IAM-chromatography experiments (n=3) revealed a P(m) value of 1.51±0.34 for Me@APPI. Stability testing using human liver microsomes revealed that 99.5% of the tracer was found to be still intact after 60 minutes (n=4). Conclusion: Present data indicate that [(11)C]Me@APPI has promising properties to become a clinically useful NET-PET-tracer. Further in-vitro and in-vivo evaluations are currently under way.
Article
An ultrafiltration system was evaluated for the free-fraction measurement of SPECT radiotracers (β-CIT, IBF, and iomazenil) used in functional brain imaging. The effect of temperature, storage, centrifugal force, tracer concentration, and percentage filtered demonstrated a relative error of < 9%. As a result of the minimal temperature effect, 25 °C was employed for all measurements. A comparison of the ultrafiltration system with equilibrium dialysis revealed < 5% difference for β-CIT and iomazenil, but 16% for IBF. Additionally, the time and ease of operation considerably favored the ultrafiltration system. The precision quantitated by repetition was < 6% for between-run and within-run variability. In conclusion, ultrafiltration provided rapid results, demonstrated minor analytical errors, revealed generally good correlation with equilibrium dialysis, and allowed excellent precision.
Article
Even in the 21st century, studies aimed at characterizing the pathological paradigms associated with the development and progression of central nervous system diseases are primarily performed in laboratory animals. However, limited translational significance, high cost, and labor to develop the appropriate model (e.g., transgenic or inbred strains) have favored parallel in vitro approaches. In vitro models are of particular interest for cerebrovascular studies of the blood-brain barrier (BBB), which plays a critical role in maintaining the brain homeostasis and neuronal functions. Because the BBB dynamically responds to many events associated with rheological and systemic impairments (e.g., hypoperfusion), including the exposure of potentially harmful xenobiotics, the development of more sophisticated artificial systems capable of replicating the vascular properties of the brain microcapillaries are becoming a major focus in basic, translational, and pharmaceutical research. In vitro BBB models are valuable and easy to use supporting tools that can precede and complement animal and human studies. In this article, we provide a detailed review and analysis of currently available in vitro BBB models ranging from static culture systems to the most advanced flow-based and three-dimensional coculture apparatus. We also discuss recent and perspective developments in this ever expanding research field.