Sei sulla pagina 1di 6

ARTICLE IN PRESS

Applied Radiation and Isotopes 67 (2009) 2019–2024

Contents lists available at ScienceDirect

Applied Radiation and Isotopes


journal homepage: www.elsevier.com/locate/apradiso

Synthesis of carbon-11 labeled celecoxib derivatives as new candidate PET


radioligands for imaging of inflammation
Mingzhang Gao a, Min Wang a, Kathy D. Miller b, Gary D. Hutchins a, Qi-Huang Zheng a,
a
Department of Radiology, Indiana University School of Medicine, 1345 West 16th Street, L3-208, Indianapolis, IN 46202, USA
b
Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA

a r t i c l e in fo abstract

Article history: Cyclooxygenase (prostaglandin endoperoxide synthase or COX) enzyme represents a particularly
Received 6 April 2009 attractive target in inflammation processes for the development of both therapeutic agents and imaging
Accepted 15 July 2009 agents. This study was designed to develop new radioligands for imaging of inflammation using the
biomedical imaging technique positron emission tomography (PET). Carbon-11 labeled celecoxib
Keywords: derivatives, [11C]methyl 2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)phenylsulfonamidooxy)
Positron emission tomography acetate ([11C]6e), [11C]methyl 2-methyl-2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)phenylsul-
Cyclooxygenase fonamidooxy)propanoate ([11C]6f), [11C]methyl 2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-
Celecoxib derivatives pyrazol-1-yl)phenylsulfonamidooxy)acetate ([11C]6g), and [11C]methyl 2-methyl-2-(4-(5-(4-methoxy-
Radioligands
phenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl)phenylsulfonamidooxy)propanoate ([11C]6h), were prepared
Inflammation
by O-[11C]methylation of their corresponding precursors using [11C]CH3OTf under basic condition and
Imaging
isolated by a simplified solid-phase extraction (SPE) method in 50–60% radiochemical yields based on
[11C]CO2 and decay corrected to end of bombardment (EOB). The overall synthesis time from EOB was
15–20 min, the radiochemical purity was 499%, and the specific activity at end of synthesis (EOS) was
111–185 GBq/mmol.
& 2009 Elsevier Ltd. All rights reserved.

1. Introduction COX inhibitors that inhibit all COX enzymes. Two major classes of
compounds have been developed as COX-2 selective inhibitors:
The enzyme cyclooxygenase (prostaglandin endoperoxide the first one is the methanesulfonamide compounds like NS-398
synthase or COX) catalyses the biosynthesis of prostaglandin, and the second is ‘‘coxib’’ family such as celecoxib (Julemont et al.,
prostacyclin and thromboxane from arachidonic acid (Anzini 2004) (Fig. 1). However, it is important to note that some COX-2
et al., 2008). Three COX isozymes including COX-1, COX-2 and selective inhibitors have been associated with significant
COX-3 are actually described (Prabhakaran et al., 2005). COX-1 is gastrointestinal side effects (Szabo et al., 2008). For example,
present in healthy tissues and responsible for the thrombogensis rofecoxib (Vioxx) has been withdrawn from the market due to the
and the homeostasis. COX-2 is almost undetectable under physio- enhanced risk for cardiovascular diseases. To improve the therapy
logic conditions and mainly induced by inflammatory stimuli. COX- with fewer side effects, recently a novel series of celecoxib
3 is located in the central nervous system and could be the derivatives have been designed and synthesized as more potent
pharmacological target of acetaminophen. COX-2 over expression anti-inflammatory agents (Szabo et al., 2008). COX-2 also provides
is associated with inflammation processes and cancer progression a particularly attractive target for the development of imaging
and contributes to the pathogenesis of several types of cancers agents, and several papers have reported the synthesis and
such as breast cancer and prostate cancer and neurodegenerative preliminary evaluation of radiolabeled COX-2 inhibitors for
diseases such as Alzheimer’s disease and Parkinson’s disease imaging of COX-2 using the biomedical imaging technique
(Kuge et al., 2006; Prabhakaran et al., 2005). COX-2 provides an positron emission tomography (PET) (de Vries et al., 2003;
attractive target for the development of therapeutic agents. It was McCarthy et al., 2002; Prabhakaran et al., 2005, 2007; Wust
proposed that a selective inhibitor of COX-2 would be a better et al., 2005). However, the potential of these radioligands for
approach to treat inflammatory conditions, since the traditional imaging COX-2 expression remains unclear. In addition, most of
non-steroidal anti-inflammatory drugs (NSAIDs) are non-selective these COX-2 radioligands are labeled with positron emitting
radionuclide fluorine-18 (de Vries et al., 2003; McCarthy et al.,
2002; Prabhakaran et al., 2007; Wust et al., 2005). Recent efforts
 Corresponding author. Tel.: +1 317 278 4671; fax: +1 317 278 9711. have turned to develop selective COX-2 radioligands labeled with
E-mail address: qzheng@iupui.edu (Q.-H. Zheng). another positron emitting radionuclide carbon-11, because some

0969-8043/$ - see front matter & 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.apradiso.2009.07.022
ARTICLE IN PRESS
2020 M. Gao et al. / Applied Radiation and Isotopes 67 (2009) 2019–2024

of fluorine-18 radioligands resulted in de[18F]fluorination in vivo contains a fairly rigid core structure with three apparent
(Prabhakaran et al., 2005, 2007). We are interested in the interaction points: R1, R2, and R3 (Fig. 1). We designed new
development of enzyme-based and/or receptor-based PET celecoxib derivatives with slight chemical modifications in the R1
imaging agents. In our previous works, we have described the and R3 positions and without any change in the R2 position. These
synthesis of carbon-11 labeled sulfonanilide analogs of NS-398 as newly designed target compounds were expected to be potent
potential PET cancer imaging agents (Wang et al., 2007). In the anti-inflammatory agents with comparable anti-inflammatory
present study, we report the design, synthesis and labeling of a activity to their parent compounds and improved metabolic
series of celecoxib derivatives, [11C]methyl 2-(4-(5-p-tolyl-3- properties, favorable lipophilicity to penetrate the blood–brain
(trifluoromethyl)-1H-pyrazol-1-yl)phenylsulfonamidooxy)acetate barrier (BBB) and O-methyl position amenable to labeling with
([11C]6e), [11C]methyl 2-methyl-2-(4-(5-p-tolyl-3-(trifluoromethyl)- carbon-11 (Ahlstrom et al., 2007; Gao et al., 2008; Liu et al., 2005;
1H-pyrazol-1-yl)phenylsulfonamidooxy)propanoate ([11C]6f), [11C] Szabo et al., 2008). Moreover, we and other groups have reported a
methyl 2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyra- methyl ester prodrug of matrix metalloproteinase (MMP) inhi-
zol-1-yl)phenylsulfonamidooxy)acetate ([11C]6g), and [11C]methyl bitor has better uptake in tumor than its acid parent drug
2-methyl-2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol- (Furumoto et al., 2003; Zheng et al., 2003). Synthesis of celecoxib
1-yl)phenylsulfonamidooxy)propanoate ([11C]6h), as new candi- derivative acid precursors and methyl ester standards was
date PET radioligands for imaging of inflammation. accomplished using a modification of the previously reported
procedures (Ahlstrom et al., 2007; Szabo et al., 2008). As depicted
in Scheme 1, Claisen condensation of the acetophenone 1 with
2. Results and discussion trifluoroacetic acid ethyl ester under strong base provided
diketone 2 in 92–93% yield. Two different bases NaH (Szabo
2.1. Chemistry et al., 2008) and NaOMe (Ahlstrom et al., 2007) were tested in
the reaction, and NaH was proved to be a better base for the
The chemistry is straightforward. Celecoxib was used as a reaction, which will give 2 at reliable high yield. Compound 2 was
model compound for the design of a novel series of celecoxib reacted with p-hydrazino-benzene-sulfonic acid 3 through a
derivatives (Ahlstrom et al., 2007; Szabo et al., 2008). Celecoxib regioselective transformation to form 1,5-diarylpyrazole 4 in
81–86% yield. The solubility of compound 3 affected the yield of
O the transformation. The yield can be improved by adding some
H 2N
R3 S water in the reaction. The intermediate 4 was transferred to
O sulfonyl chloride 5 using phosphorus pentachloride in
O 2N O N dichloromethane with catalytic amount of DMF in 86–90% yield.
N CF3 R2
Compound 5 was reacted with aminooxy-carboxylic acid
or methyl ester in dichloromethane or DMF with Et3N and
NH
4-dimethylaminopyridine (DMAP) to give its corresponding
O S O derivative 6 in 40–60% yield. The literature procedure (Szabo
CH3 et al., 2008) for the preparation of 6 gave poor yield (o30%) in our
R1 H 3C
Celecoxib hands. Therefore, we modified the literature procedure (two-
NS-398
phase reaction under dioxane/water) to a single phase reaction by
Fig. 1. COX-2 inhibitors NS-398 and celecoxib. using DMF as a solvent, Et3N as a base and DMAP as a catalyst.

O
HO
NHNH2 S
O
CF3CO2Et N
O NaH/THF, 0 oC O N CF3
HO3S 3
X X O
EtOH, H2O/HCl
CH3
1a, X=Me reflux
2a, X=Me, 93% CF3
1b, X=OMe 2b, X=OMe, 92%
X 4a, X=Me, 86%
4b, X=OMe, 81%

PCl5, CH2Cl2
O O reflux
RONH Cl
S S
O O
N N
N CF3 6a, X=Me, R=CH 2CO2H CF3
RONH2 HCl, DMF N
6b, X=Me, R=C(CH 3)2CO2H Et3N, DMAP, rt
6c, X=OMe, R=CH 2CO2H
6d, X=OMe, R=C(CH 3)2CO2H
6e, X=Me, R=CH 2CO2CH3
X 6f, X=Me, R=C(CH 3)2CO2CH3 X 5a, X=Me, 90%
6g, X=OMe, R=CH 2CO2CH3
5b, X=OMe, 86%
6h, X=OMe, R=C(CH 3)2CO2CH3
40-60%

Scheme 1. Synthesis of celecoxib derivative precursors and standards.


ARTICLE IN PRESS
M. Gao et al. / Applied Radiation and Isotopes 67 (2009) 2019–2024 2021

O O
RONH RONH
S S
O O
N
11
CH3OTf, CH3CN N
CF3 N CF3
N
2 N NaOH, 80 oC

X X
6a, X=Me, R=CH 2CO2H [11C]6e, X=Me, R=CH 2CO211CH3
6b, X=Me, R=C(CH 3)2CO2H [11C]6f, X=Me, R=C(CH 3)2CO211CH3
6c, X=OMe, R=CH 2CO2H [11C]6g, X=OMe, R=CH 2CO211CH3
6d, X=OMe, R=C(CH 3)2CO2H [11C]6h, X=OMe, R=C(CH 3)2CO211CH3
50-60%

Scheme 2. Synthesis of carbon-11 labeled celecoxib derivatives.

This modification significantly improved the yield. Compounds 497%. The radiochemical purity of the target tracers was 499%
6a–d are acid precursors for carbon-11 labeling, and compound determined by radio-HPLC through g-ray (PIN diode) flow
6e–h are methyl ester reference standards. detector, and the chemical purity of the target tracers was
495% determined by reversed-phase HPLC through UV flow
detector.
2.2. Radiochemistry

Synthesis of carbon-11 labeled celecoxib derivatives [11C]6e–h


is indicated in Scheme 2. Acid precursors 6a–d were labeled by a 3. Materials and methods
reactive [11C]methylating agent, [11C]methyl triflate ([11C]CH3OTf)
(Jewett, 1992; Mock et al., 1999) prepared from [11C]CO2, 3.1. General
under basic conditions (2 N NaOH) in acetonitrile through the
O-[11C]methylation and isolated by a simplified solid-phase All commercial reagents and solvents from Aldrich and Sigma
extraction (SPE) method (Yoder et al., 2009; Zheng and were used without further purification. [11C]CH3OTf was prepared
Mulholland, 1996) to provide target tracers [11C]6e–h in 50–60% according to a literature procedure (Mock et al., 1999). Melting
radiochemical yields, decay corrected to end of bombardment points were determined on a MEL-TEMP II capillary tube
(EOB), based on [11C]CO2. The large polarity difference between apparatus and were uncorrected. 1H NMR spectra were recorded
the acid precursor and the labeled O-methylated ester product on Varian Gemini 2000 200 MHz FT-NMR and Bruker Avance II
permitted the use of SPE technique for purification of the labeled 500 MHz NMR spectrometers using tetramethylsilane (TMS) as an
product from the radiolabeling reaction mixture. Either a C-18 internal standard. Chemical shift data for the proton resonances
Plus Sep-Pak cartridge (disposable) or a semi-prep C-18 guard were reported in parts per million (ppm, d scale) relative to
cartridge column (repeat use) was used in SPE purification internal standard TMS (d 0.0), and coupling constants (J) were
technique. The crude reaction mixture was treated with sodium reported in hertz (Hz). Low resolution mass spectra (LRMS) were
bicarbonate and loaded onto the cartridge by gas pressure. Any obtained using a Bruker Biflex III MALDI-Tof mass spectrometer,
non-reacted acid precursor was actually converted to the and high resolution mass spectra (HRMS) were obtained using a
corresponding sodium salt, and any non-reacted [11C]CH3OTf Thermo MAT 95XP-Trap spectrometer. Chromatographic solvent
was actually hydrolyzed to [11C]CH3OH, which would not stick to proportions are indicated as volume: volume ratio. Thin-layer
the C-18 Sep-Pak. The cartridge was washed with water to remove chromatography (TLC) was run using Analtech silica gel GF
non-reacted [11C]CH3OTf, acid precursor and reaction solvent, and uniplates (5  10 cm2). Plates were visualized under UV light.
then the final labeled product was eluted with ethanol. Overall Preparative TLC was run using Analtech silica gel UV 254 plates
synthesis time was 15–20 min from EOB. SPE technique is fast, (20  20 cm2). Normal phase flash column chromatography was
efficient and convenient and works very well for the O-methylated carried out on EM Science silica gel 60 (230–400 mesh) with a
ester tracer purification using the acid precursor for radiolabel- forced flow of the indicated solvent system in the proportions
ing (Yoder et al., 2009; Zheng and Mulholland, 1996). The described below. All moisture- and/or air-sensitive reactions were
radiosynthesis was performed in an automated multi-purpose performed under a positive pressure of nitrogen maintained by a
11
C-radiosynthesis module, allowing measurement of specific direct line from a nitrogen source.
activity during synthesis (Mock et al., 2005a, b). The specific Analytical HPLC was performed using a Prodigy (Phenomenex)
activity was estimated in a range of 111–185 GBq/mmol at the end 5 mm C-18 column, 4.6  250 mm; 3:1:1 CH3CN:MeOH:20 mM, pH
of synthesis (EOS) based on other compounds produced using the 6.7 phosphate (buffer solution) mobile phase; flow rate 1.5 mL/
same targetry conditions which have been measured by the on- min; and UV (254 nm) and g-ray (PIN diode) flow detectors. C-18
the-fly technique (Mock et al., 2005a; Zheng et al., 2007). The Plus Sep-Pak cartridges were obtained from Waters Corporate
actual measurement of specific activity at EOS by analytical HPLC Headquarters, Milford, MA. Semi-prep C-18 guard cartridge
method (Zheng and Mock, 2005) is in agreement with this column 1 1 cm was obtained from E.S. Industries, Berlin, NJ,
estimation. Chemical purity and radiochemical purity were and part number 300121-C18-BD 10 mm. Sterile Millex-GS
determined by analytical HPLC method (Zheng and Mock, 2005). 0.22 mm vented filter unit was obtained from Millipore Corpora-
The chemical purity of the precursors and reference standards was tion, Bedford, MA.
ARTICLE IN PRESS
2022 M. Gao et al. / Applied Radiation and Isotopes 67 (2009) 2019–2024

3.2. 4,4,4-Trifluoro-1-(4-methylphenyl)butane-1,3-dione (2a) residue was crystallized from hexane and ethyl acetate mixture to
give 5a (2.38 g, 90%) as a white solid, mp 85–87 1C. 1H NMR
4-Methyl-acetophenone 1a (20.0 g, 149 mmol) was dissolved in (CDCl3): d 2.40 (s, 3H, CH3), 6.75 (s, 1H, CH ¼ ), 7.10–7.25 (m, 4H,
dry THF (300 mL) under nitrogen atmosphere and NaH (7.15 g, Ph-H), 7.56 (d, J ¼ 8.8 Hz, 2H, Ph-H), 7.99 (d, J ¼ 8.8 Hz, 2H, Ph-H).
298 mmol) was added in portions maintaining the temperature
between 5 and 0 1C. After stirring at this temperature for 30 min, 3.7. 4-(5-(4-Methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-
ethyl trifluoroacetate (31.75 g, 224 mmol) was added and the yl)benzene-1-sulfonyl chloride (5b)
reaction mixture was allowed to stir at room temperature (rt) for
6 h. The reaction mixture was evaporated, added with ice-water,
Compound 5b was prepared from 4b using the procedure
acidified with HCl (1 N) to pH 6, and extracted with ethyl acetate
described for 5a as a white solid in 86% yield, mp 109–110 1C. 1H
(3  100 mL). The combined organic layer was washed with water
NMR (CDCl3): d 3.85 (s, 3H, CH3), 6.73 (s, 1H, CH ¼ ), 6.90
(50 mL), dried over MgSO4, and concentrated under reduced
(d, J ¼ 8.8 Hz, 2H, Ph-H), 7.15 (d, J ¼ 8.6 Hz, 2H, Ph-H), 7.56
pressure. The solid residue was washed with hexane and dried
(d, J ¼ 8.8 Hz, 2H, Ph-H), 7.99 (d, J ¼ 8.8 Hz, 2H, Ph-H).
under high vacuum to provide a solid mass, which was re-
dissolved in dichloromethane and dried under high vacuum to
give 2a (31.87 g, 93%) as a light brown solid, Rf ¼ 0.25 (25% EtOAc/ 3.8. General procedure for preparation of celecoxib derivative 6
hexanes), mp 40–42 1C. 1H NMR (CDCl3): d 2.44 (s, 3H, CH3), 6.54
(s, 1H, CH ¼ ), 7.28 (d, J ¼ 8.0 Hz, 2H, Ph-H), 7.83 (d, J ¼ 8.0 Hz, 2H, The benzenesulfonyl chloride compound 5 (0.5 mmol) was
Ph-H). MS (EI): 229 ([M–H]+, 91%), 159 (100%). added to a solution of aminooxy-carboxylic acid hydrochloride or
aminooxy-carboxylic acid methyl ester (0.6 mmol) and Et3N
3.3. 4,4,4-Trifluoro-1-(4-methoxyphenyl)butane-1,3-dione (2b) (101 mg, 1.0 mmol) in dichloromethane (40 mL). DMAP was
added to this solution (61 mg, 0.5 mmol) and stirred at rt for 5 h.
Compound 2b was prepared from 4-methoxy-acetophenone Then the reaction mixture was concentrated under vacuum. The
1b using the procedure described for 2a as a light brown solid in residue was added with water and extracted with ethyl acetate
92% yield, Rf ¼ 0.24 (25% EtOAc/hexanes), mp 48–49 1C. 1H NMR (2  80 mL), dried over MgSO4, evaporated, and purified by
(CDCl3): d 3.90 (s, 3H, OCH3), 6.50 (s, 1H, CH ¼ ), 6.96 column chromatography on silica gel (5% MeOH/CH2Cl2) or (10%
(d, J ¼ 8.8 Hz, 2H, Ph-H), 7.91 (d, J ¼ 9.0 Hz, 2H, Ph-H). MS (ESI): EtOAc/hexanes) to afford 6 as a white solid in 40–60% yield.
247 ([M+H]+, 100%). 2-(4-(5-p-Tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)phenylsu-
lfonamidooxy)acetic acid (6a), Rf ¼ 0.73 (20% MeOH/CH2Cl2), mp
221–223 1C. 1H NMR (CDCl3): d 2.31 (s, 3H, CH3), 4.37 (s, 2H, CH2),
3.4. 4-(5-p-Tolyl-3-(trifluoromethyl)-1H-pyrazol-1-
6.96 (s, 1H, CH ¼ ), 7.20–7.24 (m, 4H, Ph-H), 7.56–7.57 (m, 2H,
yl)benzenesulfonic acid (4a)
Ph-H), 7.95–7.96 (m, 2H, Ph-H), 9.9 (s, 1H). MS (ESI): 456 ([M+H]+,
100%).
Compound 2a (6.00 g, 26 mmol) was added into a solution of p-
2-Methyl-2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)
hydrazino-benzenesulfonic acid 3 (5.64 g, 30 mmol) in ethanol
phenylsulfonamidooxy)propanoic acid (6b), Rf ¼ 0.80 (20% MeOH/
(320 mL) and HCl (6 N, 8.6 mL) and water (40 mL). The mixture
CH2Cl2), mp 93–95 1C. 1H NMR (CDCl3): d 1.33 (s, 6H, 2xCH3),
was heated to reflux and stirred for 20 h. Then the reaction
2.35 (s, 3H, CH3), 6.72 (s, 1H, CH ¼ ), 7.06 (d, J ¼ 7.5 Hz, 2H, Ph-H),
mixture was evaporated under reduced pressure, extracted with
7.14 (d, J ¼ 7.5 Hz, 2H, Ph-H), 7.43 (d, J ¼ 7.5 Hz, 2H, Ph-H), 7.85
ethyl acetate (4  120 mL), washed with brine, dried over MgSO4,
(d, J ¼ 8.0 Hz, 2H, Ph-H), 8.01 (s, 1H). MS (ESI): 484 ([M+H]+,
filtered, and evaporated to dryness. The residue was purified by
100%).
column chromatography on silica gel (12% MeOH/CH2Cl2) to
2-(4-(5-(4-Methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-
afford 4a (8.54 g, 86%) as a white solid, Rf ¼ 0.14 (10% MeOH/
yl)phenylsulfonamidooxy)acetic acid (6c), Rf ¼ 0.68 (20% MeOH/
CH2Cl2), mp4280 1C. 1H NMR (DMSO-d6): d 2.30 (s, 3H, CH3), 7.14
CH2Cl2), mp 108–110 1C. 1H NMR (CDCl3): d 3.75 (s, 3H, CH3), 4.30
(s, 1H, CH ¼ ), 7.18–7.19 (m, 4H, Ph-H), 7.26 (d, J ¼ 8.8 Hz, 2H, Ph-
(s, 2H, CH2), 4.39 (s, 1H), 6.87 (s, 1H, CH ¼ ), 6.91 (d, J ¼ 7.2 Hz, 2H,
H), 7.62 (d, J ¼ 8.8 Hz, 2H, Ph-H). MS (ESI): 383 ([M+H]+, 100%).
Ph-H), 7.20 (d, J ¼ 8.2 Hz, 2H, Ph-H), 7.53 (d, J ¼ 8.2 Hz, 2H, Ph-H),
7.89 (d, J ¼ 7.2 Hz, 2H, Ph-H). MS (ESI): 472 ([M+H]+, 100%). HRMS
3.5. 4-(5-(4-Methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-1- (CI): calcd for C19H17O6N3F3S1 ([M+H]+), 472.0785; found
yl)benzenesulfonic acid (4b) 472.0767.
2-(4-(5-(4-Methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-
Compound 4b was prepared from 2b using the procedure yl)phenylsulfonamidooxy)-2-methylpropanoic acid (6d), Rf ¼ 0.77
described for 4a as a white solid in 81% yield, Rf ¼ 0.12 (10% (20% MeOH/CH2Cl2), mp 97–99 1C. 1H NMR (DMSO-d6): d 1.47
MeOH/CH2Cl2), mp 298–300 1C. 1H NMR (DMSO-d6): d 3.76 (s, 3H, (s, 6H, 2  CH3), 3.83 (s, 3H, CH3), 6.78 (s, 1H, CH ¼ ), 6.90
CH3), 6.94 (d, J ¼ 8.8 Hz, 2H, Ph-H), 7.10 (s, 1H, CH ¼ ), 7.20 (d, J ¼ 8.0 Hz, 2H, Ph-H), 7.19 (d, J ¼ 8.5 Hz, 2H, Ph-H), 7.51
(d, J ¼ 9.0 Hz, 2H, Ph-H), 7.24 (d, J ¼ 8.6 Hz, 2H, Ph-H), 7.63 (d, J ¼ 8.0 Hz, 2H, Ph-H), 7.65 (s, 1H), 7.94 (d, J ¼ 8.0 Hz, 2H, Ph-H),
(d, J ¼ 8.4 Hz, 2H, Ph-H). MS (ESI): 399 ([M+H]+, 100%). 8.82 (s, 1H). MS (ESI): 499 (M+, 100%). HRMS (ESI): calcd for
C21H21O6N3F3S1 ([M+H]+), 500.1103; found 500.1081.
3.6. 4-(5-p-Tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)benzene-1- Methyl 2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1-yl)-
sulfonyl chloride (5a) phenylsulfonamidooxy)acetate (6e), Rf ¼ 0.67 (34% EtOAc/hex-
anes), mp 137–139 1C. 1H NMR (CDCl3): d 2.39 (s, 3H, CH3), 3.81
Phosphorus pentachloride (2.2 g, 10.5 mmol) was added part- (s, 3H, CH3), 4.51 (s, 2H, CH2), 6.74 (s, 1H, CH ¼ ), 7.11 (d, J ¼ 8.0 Hz,
wise to a suspension of compound 4a (2.52 g, 6.6 mmol) in 2H, Ph-H), 7.18 (d, J ¼ 8.0 Hz, 2H, Ph-H), 7.48 (d, J ¼ 9.0 Hz, 2H,
dichloromethane (60 mL). Then DMF (1 mL) was added to the Ph-H), 7.79 (s, 1H), 7.89 (d, J ¼ 9.0 Hz, 2H, Ph-H). MS (ESI): 470
reaction solution. The resulting solution was refluxed for 8 h, ([M+H]+, 100%). HRMS (ESI): calcd for C20H19O5N3F3S1 ([M+H]+),
and the mixture was concentrated under vacuum. Ice-water 470.0998; found 470.1003.
was added to the residue and extracted with ethyl acetate Methyl 2-methyl-2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyra-
(2  120 mL), dried over MgSO4, and evaporated to dryness. The zol-1-yl)phenylsulfonamidooxy)propanoate (6f), Rf ¼ 0.71 (34%
ARTICLE IN PRESS
M. Gao et al. / Applied Radiation and Isotopes 67 (2009) 2019–2024 2023

EtOAc/hexanes), mp 53–55 1C. 1H NMR (CDCl3): d 1.46 (s, 6H, 6c ¼ 3.14 min, tR 6g ¼ 5.56 min, tR [11C]6g ¼ 5.56 min; and tR
2  CH3), 2.38 (s, 3H, CH3), 3.72 (s, 3H, CH3), 6.74 (s, 1H, CH ¼ ), 6d ¼ 3.25 min, tR 6h ¼ 5.47 min, tR [11C]6h ¼ 5.47 min.
7.09 (d, J ¼ 8.0 Hz, 2H, Ph-H), 7.16 (d, J ¼ 8.4 Hz, 2H, Ph-H), 7.33
(s, 1H), 7.48 (d, J ¼ 8.8 Hz, 2H, Ph-H), 7.89 (d, J ¼ 8.8 Hz, 2H, Ph-H).
MS (ESI): 498 ([M+H]+, 100%). HRMS (ESI): calcd for C22H22O5N3- 4. Conclusions
F3S1Na ([M+Na]+), 520.1130; found 520.1104.
Methyl 2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H- An efficient and convenient synthesis of new radioligands,
pyrazol-1-yl)phenylsulfonamidooxy)acetate (6g), Rf ¼ 0.53 (34% carbon-11 labeled celecoxib derivatives, has been well developed.
EtOAc/hexanes), mp 106–108 1C. 1H NMR (CDCl3): d 3.76 (s, 3H, The synthetic methodology employed classical organic chemistry
CH3), 3.83 (s, 3H, CH3), 4.51 (s, 2H, CH2), 6.71 (s, 1H, CH ¼ ), 6.87 such as Claisen condensation, regioselective transformation,
(d, J ¼ 8.8 Hz, 2H, Ph-H), 7.13 (d, J ¼ 8.4 Hz, 2H, Ph-H), 7.49 sulfonylation and amidating reaction to prepare a series of new
(d, J ¼ 8.8 Hz, 2H, Ph-H), 7.80 (s, 1H), 7.89 (d, J ¼ 8.6 Hz, 2H, Ph-H). celecoxib derivative precursors and standard compounds. The
MS (ESI): 486 ([M+H]+, 100%). HRMS (ESI): calcd for C20H19O6N3 target radioligands were prepared by O-[11C]methylation of their
F3S1 ([M+H]+), 486.0947; found 486.0957. corresponding acid precursors using a reactive [11C]methylating
Methyl 2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H- agent, [11C]CH3OTf, and isolated by a simplified SPE purification
pyrazol-1-yl)phenylsulfonamidooxy)-2-methylpropanoate (6h), procedure in high radiochemical yields, short overall synthesis
Rf ¼ 0.60 (34% EtOAc/hexanes), mp 80–82 1C. 1H NMR (CDCl3): d time, and great specific radioactivities. These chemistry results
1.46 (s, 6H, 2  CH3), 3.72 (s, 3H, CH3), 3.83 (s, 3H, CH3), 6.72 combined with the reported in vitro and in vivo biological data
(s, 1H, CH ¼ ), 6.87 (d, J ¼ 8.8 Hz, 2H, Ph-H), 7.13 (d, J ¼ 8.8 Hz, (Ahlstrom et al., 2007; Prabhakaran et al., 2005, 2007; Szabo et al.,
2H, Ph-H), 7.33 (s, 1H), 7.49 (d, J ¼ 8.8 Hz, 2H, Ph-H), 7.88 2008) encourage further in vivo biological evaluation of new
(d, J ¼ 8.6 Hz, 2H, Ph-H). MS (ESI): 514 ([M+H]+, 100%). HRMS carbon-11 labeled celecoxib derivatives, as candidate radioligands
(ESI): calcd for C22H23O6N3F3S1 ([M+H]+), 514.1260; found to image inflammation.
514.1254.

Acknowledgments
3.9. General procedure for preparation of target tracers, [11C]methyl
2-(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1- This work was partially supported by the Susan G. Komen for
yl)phenylsulfonamidooxy)acetate ð½11 C6eÞ, [11C]methyl 2-methyl-2- the Cure, Breast Cancer Research Foundation and Indiana
(4-(5-p-tolyl-3-(trifluoromethyl)-1H-pyrazol-1- Genomics Initiative (INGEN) of Indiana University, which is
yl)phenylsulfonamidooxy)propanoate ð½11 C6fÞ, [11C]methyl 2-(4-(5- supported in part by Lilly Endowment Inc. The authors would
(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-1- like to thank Dr. Bruce H. Mock and Barbara E. Glick-Wilson for
yl)phenylsulfonamidooxy)acetate ð½11 C6gÞ, and [11C]methyl 2- their assistance in production of [11C]CH3OTf.
methyl-2-(4-(5-(4-methoxyphenyl)-3-(trifluoromethyl)-1H-pyrazol-
1-yl)phenylsulfonamidooxy)propanoate ð½11 C6hÞ References
11 14 11
[ C]CO2 was produced by the N(p,a) C nuclear reaction in Ahlstrom, M.M., Ridderstrom, M., Zamora, I., Luthman, K., 2007. CYP2C9 structure-
small volume (9.5 cm3) aluminum gas target (CTI) from 11 MeV metabolism relationships: optimizing the metabolic stability of COX-2
proton cyclotron on research purity nitrogen (+1% O2) in a inhibitors. J. Med. Chem. 50, 4444–4452.
Anzini, M., Rovin, M., Cappeli, A., Vomero, S., Manetti, F., Botta, M., Sautebin, L.,
Siemens radionuclide delivery system (Eclipse RDS-111). The acid Rossi, A., Pergola, C., Ghelardini, C., Norcini, M., Giordani, A., Makovec, F.,
precursor 6a, 6b, 6c, or 6d (0.1–0.3 mg) was dissolved in CH3CN Anzellotti, P., Patrignani, P., Biava, M., 2008. Synthesis, biological evaluation,
(300 mL). To this solution was added 2 N NaOH (2 mL). The mixture and enzyme docking simulations of 1,5-diarylpyrrole-3-alkoxyethyl ethers as
selective cyclooxygenase-2 inhibitors endowed with anti-inflammatory and
was transferred to a small reaction vial. No-carrier-added (high antinociceptive activity. J. Med. Chem. 51, 4476–4481.
specific activity) [11C]CH3OTf that was produced by the gas-phase de Vries, E.F., van Waarde, A., Buursma, A.R., Vaalburg, W., 2003. Synthesis and in
production method (Mock et al., 1999) from [11C]CO2 through vivo evaluation of 18F-desbromo-DuP-697 as a PET tracer for cyclooxygenase-2
expression. J. Nucl. Med. 44, 1700–1706.
[11C]CH4 and [11C]CH3Br with silver triflate (AgOTf) column was
Furumoto, S., Takashima, K., Kubota, K., Ido, T., Iwata, R., Fukuda, H., 2003. Tumor
passed into the reaction vial at rt until radioactivity reached a detection using 18F-labeled matrix metalloproteinase-2 inhibitor. Nucl. Med.
maximum (2 min), and then the reaction vial was isolated and Biol. 30, 119–125.
heated at 80 1C for 3 min. The contents of the reaction vial were Gao, M., Wang, M., Hutchins, G.D., Zheng, Q.-H., 2008. Synthesis of new carbon-11
labeled benzoxazole derivatives for PET imaging of 5-HT3 receptor. Eur. J. Med.
diluted with NaHCO3 (0.1 M, 1 mL). The reaction tube was Chem. 43, 1570–1574.
connected to either a C-18 Plus Sep-Pak cartridge or a semi-prep Jewett, D.M., 1992. A simple synthesis of [11C]methyl triflate. Int. J. Rad. Appl.
C-18 guard cartridge column. The labeled product mixture Instrum. A 43, 1383–1385.
Julemont, F., de Leval, X., Michaux, C., Renard, J.-F., Winum, J.-Y., Montero, J.-L.,
solution was passed onto the cartridge for SPE purification by Damas, J., Dogne, J.-M., Pirotte, B., 2004. Design, synthesis, and pharmacolo-
gas pressure. The cartridge was washed with H2O (2  3 mL), and gical evaluation of pyridinic analogues of nimesulide as cyclooxygenase-2
the aqueous washing was discarded. The product was eluted from selective inhibitors. J. Med. Chem. 47, 6749–6759.
Kuge, Y., Katada, Y., Shimonaka, S., Temma, T., Kimura, H., Kiyono, Y., Yokota, C.,
the column with EtOH (2  3 mL), and then passed onto a rotatory Minematsu, K., Seki, K.-i., Tamaki, T., Ohkura, K., Saji, H., 2006. Synthesis and
evaporator. The solvent was removed by evaporation under evaluation of radioiodinated cyclooxygenase-2 inhibitors as potential SPECT
vacuum. The labeled product [11C]6e, [11C]6f, [11C]6g, or [11C]6h tracers for cyclooxygenase-2 expression. Nucl. Med. Biol. 33, 21–27.
Liu, X., Wang, J.-Q., Zheng, Q.-H., 2005. Lipophilicity coefficients of potential tumor
was formulated with saline, sterile-filtered through a sterile imaging agents, positron-labeled O6-benzylguanine derivatives. Biomed.
vented Millex-GS 0.22 mm cellulose acetate membrane and Chromatogr. 19, 379–384.
collected into a sterile vial. Total radioactivity was assayed and McCarthy, T.J., Sheriff, A.U., Graneto, M.J., Talley, J.J., Welch, M.J., 2002. Radio-
synthesis, in vitro validation, and in vivo evaluation of 18F-labeled COX-1 and
the total volume was noted for tracer dose dispensing. The overall
COX-2 inhibitors. J. Nucl. Med. 43, 117–124.
synthesis time including SPE purification and formulation was Mock, B.H., Mulholland, G.K., Vavrek, M.T., 1999. Convenient gas phase bromina-
15–20 min. The radiochemical yields decay corrected to EOB, from tion of [11C]methane and production of [11C]methyl triflate. Nucl. Med. Biol. 26,
[11C]CO2, were 50–60%. Retention times in the analytical HPLC 467–471.
Mock, B.H., Glick-Wilson, B.E., Zheng, Q.-H., DeGrado, T.R., 2005a. Automated
system were: tR 6a ¼ 2.83 min, tR 6e ¼ 4.68 min, tR [11C]6e ¼ 4.68 measurement of specific activity of radiolabeled ligands during synthesis. J.
min; tR 6b ¼ 2.75 min, tR 6f ¼ 4.95 min, tR [11C]6f ¼ 4.95 min; tR Labelled Compd. Radiopharm. 48, S224.
ARTICLE IN PRESS
2024 M. Gao et al. / Applied Radiation and Isotopes 67 (2009) 2019–2024

Mock, B.H., Zheng, Q.-H., DeGrado, T.R., 2005b. A multi-purpose 11C-radio- Yoder, K.K., Hutchins, G.D., Mock, B.H., Fei, X., Winkle, W.L., Gitter, B.D., Territo, P.R.,
synthesis system. J. Labelled Compd. Radiopharm. 48, S225. Zheng, Q.-H., 2009. Dopamine transporter binding in rat striatum: a
Prabhakaran, J., Majo, V.J., Simpson, N.S., Van Heertum, R.L., Monn, J.J., Kumar, comparison of [O-methyl-11C]b-CFT and [N-methyl-11C]b-CFT. Nucl. Med. Biol.
J.S.D., 2005. Synthesis of [11C]celecoxib: a potential PET probe for imaging COX- 36, 11–16.
2 expression. J. Labelled Compd. Radiopharm. 48, 887–895. Zheng, Q.-H., Mulholland, G.K., 1996. Improved synthesis of b-CIT and [11C]b-CIT
Prabhakaran, J., Underwood, M.D., Parsey, R.V., Arango, V., Majo, V.J., Simpson, N.R., labeled at nitrogen or oxygen positions. Nucl. Med. Biol. 23, 981–986.
Van Heertum, R., Mann, J.J., Kumar, J.S.D., 2007. Synthesis and in vivo Zheng, Q.-H., Fei, X., DeGrado, T.R., Wang, J.-Q., Stone, K.L., Martinez, T.D., Gay, D.J.,
evaluation of [18F]-4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1- Baity, W.L., Mock, B.H., Glick-Wilson, B.E., Sullivan, M.L., Miller, K.D., Sledge,
yl]benzenesulfonamide as a PET imaging probe for COX-2 expression. Bioorg. G.W., Hutchins, G.D., 2003. Synthesis, biodistribution and micro-PET imaging
Med. Chem. 15, 1802–1807. of a potential cancer biomarker carbon-11 labeled MMP inhibitor (2R)-2-[[4-
Szabo, G., Fischer, J., Kis-Varga, A., Gyires, K., 2008. New celecoxib derivatives as (6-fluorohex-1-ynyl)phenyl]sulfonylamino]-3-methylbutyric acid [11C]methyl
anti-inflammatory agents. J. Med. Chem. 51, 142–147. ester. Nucl. Med. Biol. 30, 753–760.
Wang, M., Lacy, G., Gao, M., Miller, K.D., Sledge, G.W., Zheng, Q.-H., 2007. Zheng, Q.-H., Mock, B.H., 2005. Purification of carbon-11 PET radiotracers from
Synthesis of carbon-11 labeled sulfonanilide analogues as new potential PET unlabeled precursors by preparative HPLC and SPE. Biomed. Chromatogr. 19,
agents for imaging of aromatase in breast cancer. Bioorg. Med. Chem. Lett. 17, 671–676.
332–336. Zheng, Q.-H., Gao, M., Mock, B.H., Wang, S., Hara, T., Nazih, R., Miller, M.A.,
Wust, F.R., Hohne, A., Metz, P., 2005. Synthesis of 18F-labelled cyclooxygenase-2 Receveur, T.J., Lopshire, J.C., Groh, W.J., Zipes, Z.P., Hutchins, G.D., DeGrado, T.R.,
(COX-2) inhibitors via Stille reaction with 4-[18F]fluoroiodobenzene as radio- 2007. Synthesis and biodistribution of new radiolabeled high-affinity choline
tracers for positron emission tomography (PET). Org. Biomol. Chem. 3, transporter inhibitors [11C]hemicholinium-3 and [18F]hemicholinium-3.
503–507. Bioorg. Med. Chem. Lett. 17, 2220–2224.

Potrebbero piacerti anche