• Non ci sono risultati.

9 Radiohalogens for PET Imaging

N/A
N/A
Protected

Academic year: 2021

Condividi "9 Radiohalogens for PET Imaging"

Copied!
20
0
0

Testo completo

(1)

9 Radiohalogens for PET Imaging

N Scott Mason and Chester A Mathis

Introduction

The radiohalogens are particularly attractive to con- sider as radiolabels for positron emission tomography (PET) radiophamaceuticals. While positron-emitting chlorine has not been utilized, there are several ra- dioiodines and radiobromines and one radiofluorine of great importance to PET (Table 9.1). The chemistry of radioiodine and radiobromine are similar in many re- spects, but the chemistry of radiofluorine (i.e., fluorine-18) is sufficiently unique to warrant consider- able discussion [1]. The emphasis in this chapter is upon fluorine-18 chemistry and

18

F-labeled radiophar- maceuticals. This is because

18

F, in the form of 2-deoxy- 2-[

18

F]fluoro-D-glucose (FDG), has become the most utilized PET radionuclide. Several positron-emitting radiobromines and radioiodines are not included in Table 9.1, as there is little literature regarding their routine production and use in PET imaging. While

75

Br [2, 3] and

122

I [4, 5] are included in Table 9.1, they are

not discussed further in this chapter. The half-life of

75

Br is close to that of

18

F, and its production and purification are more complicated. The short half-life of

122

I can be an advantage for blood-flow studies, but production constraints have required the use of a high- energy cyclotron and this has limited its application as well.

In general, radioiodines, radiobromines, and fluorine-18 can react as electrophiles or nucleophiles involving species that behave formally as positively charged (X

+

) or negatively charged (X

) ions, respec- tively (Figs. 9.1 and 9.2). As the names imply, elec- trophiles are electron-deficient species that seek electron-rich reactants such as carbon atoms with high local electron densities, and nucleophiles are electron- rich species that seek electron-deficient reactants [6].

While free-radical radiohalogen labeling reactions have been utilized, they tend to be disfavored for PET radiolabeling applications as a result of the difficulty in controlling the regioselectivity of this type of reaction.

In contrast, electrophilic and nucleophilic radioiodina-

203 Table 99.1. Cyclotron-produced PET radiohalogens

Radionuclide Half-life Decay Modes (%) Max. β+ Energy (MeV) Production Reactions

18

F 109.8 min β+ (97) EC (3) 0.635

18

O(p,n)

18

F

20

Ne(d, α)

18

F

76

Br 16.1 h β+ (57) EC (43) 3.98

75

As(

3

He,2n)

76

Br

76

Se(p,n)

76

Br

75

Br 98 min β+ (76) EC (24) 1.74

75

As(

3

He,3n)

75

Br

78

Kr(p, α)

75

Br

124

I 4.2 d β+ (25) EC (75) 2.13

124

Te(p,n)

124

I

124

Te(d,2n)

124

I

122

I 3.6 min β+ (77) EC (23) 3.12

127

I(p,6n)

122

Xe/

122

I

*

Chapter reproduced from Valk PE, Bailey DL, Townsend DW, Maisey MN. Positron Emission Tomography: Basic Science and

Clinical Practice. Springer-Verlag London Ltd 2003, 217–236.

(2)

tions and radiobrominations are more readily con- trolled and can be regiospecific in many cases. While nucleophilic reactions involving [

18

F]fluoride can be regiospecific as well, electrophilic [

18

F]fluorine is very reactive, and its reactions are more difficult to control.

Electrophilic fluorinations require special methods, ra- diolabeling precursors, and “taming” reagents that are described in this chapter.

Radiohalogen Production

Cyclotrons for PET Radionuclide Production

Cyclotrons have been utilized for the production of medical radionuclides since the 1930s, and cyclotron

acceleration of charged particles remains the preferred method to produce short-lived positron-emitting ra- dionuclides. While a variety of different size and energy cyclotrons have been employed for these pur- poses over the past 70 years, the brief discussion pre- sented here will be limited to an overview of the most common PET cyclotrons in operation (Table 9.2). More complete discussions of cyclotrons that produce ra- dionuclides can be found in several reviews [7–9]. Over the past ten years, most PET radionuclide production sites have installed one of two types of cyclotrons. In many academic medical settings and dedicated

18

F- production sites (particularly in the USA), the type of cyclotron most commonly in use is a single-particle cyclotron that produces protons with kinetic energies of about 11 MeV. This type of cyclotron allows access to the four most commonly utilized PET radionuclides (fluorine-18, carbon-11, nitrogen-13, and oxygen-15).

Improvements in targetry design and increases in Figure 99.1. Nucleophilic reactions relevant to [

18

F]fluoride.

Figure 99.2. Electrophilic reactions relevant to [

18

F]fluorine.

(3)

available beam currents have made these machines capable of producing multi-Curie (>74 GBq) quantities of the four common PET radionuclides. Low-energy, proton-only cyclotrons are commercially available from several suppliers, and many are available as self- shielded models. This feature helps to simplify site selection in existing building space.

The second most utilized cyclotrons (particularly in larger research-oriented facilities) are dual particle (proton (p) and deuteron (d)) 18 MeV accelerators.

These machines can produce usable quantities of other PET radionuclides, such as

76

Br,

124

I, and

64

Cu, in addi- tion to the four common PET radionuclides mentioned above. These cyclotrons also utilize less expensive, nat- urally abundant [

14

N]nitrogen to produce [

15

O]oxygen.

Production of Fluorine-18

The most common method utilized to produce nucle- ophilic [

18

F]fluoride is the

18

O(p,n)

18

F nuclear reaction (indicating the reaction of an accelerated proton (p) with oxygen-18 to produce a neutron (n) and fluorine- 18). The oxygen-18 target material most frequently consists of highly enriched [

18

O]water [10–12], but [

18

O]oxygen gas has been used successfully for this purpose as well [13]. Multi-Curie (>74 GBq) quantities of high-specific-activity [

18

F]fluoride can be produced in a few hours using 11 MeV protons to irradiate [

18

O]water targets. In addition, the separation and re- covery of [

18

O]water target material from [

18

F]fluoride is possible [14–16]. While the theoretical specific activ- ity of carrier-free

18

F is 1.7 × 10

6

Ci/mmol (6.3 × 10

7

GBq/mmol), the no-carrier-added specific activity of [

18

F]fluoride produced from [

18

O]water targets has been in the range of about 1 × 10

4

Ci/mmol (3.7 × 10

5

GBq/mmol). Other nuclear reaction pathways such

as

16

O(

3

He,p)

18

F,

16

O(

4

He,pn)

18

F, and [

6

Li(n, α)

3

H,

16

O(

3

H,n)

18

F] have been utilized in the past [17–21] but are not the current method of choice.

Two processes are currently employed to generate electrophilic [

18

F]fluorine gas ([

18

F]F

2

). One method utilizes deuteron bombardment of neon-20 gas target material to produce

18

F by the

20

Ne(d,α)

18

F reaction. A passivated nickel target (NiF) is loaded with 0.1%

(cold) F

2

in neon-20 and irradiated with 8–18 MeV deuterons. This method produces relatively low specific activity [

18

F]F

2

(~12 Ci/mmol or ~444 GBq/mmol) [22], dependent upon total mass of added carrier F

2

. The “double-shoot” method also uses a passivated nickel target; however, the target is loaded with [

18

O]oxygen gas. Proton irradiation of the target leads to adherence of radioactive

18

F species on the target walls. Upon cryogenic removal of the [

18

O]oxygen from the target, (cold) fluorine (1.0%; 30–70 μmol) diluted in a suitable inert carrier gas (for example, argon) is added to the target. A second short irradiation leads to the interaction of the carrier fluorine and surface- bound

18

F to yield recoverable [

18

F]F

2

[23,24]. Both of these methods produce relatively low yields of [

18

F]F

2

(<1 Ci or <37 GBq), and the specific activity of [

18

F]F

2

is low compared to that achievable using [

18

O]water/[

18

F]fluoride target technology.

In general, electrophilic

18

F-fluorination reactions have resulted in low-specific-activity products (<1 GBq/ μmol). A multi-step method used to produce considerably higher-specific-activity [

18

F]F

2

(>50 GBq/μmol) is worthy of note. This method utilized the (p,n) reaction on [

18

O]water as the starting point, as opposed to deuteron irradiation of neon-20 or proton irradiation of [

18

O]oxygen gas [24]. Standard proton irradiation of an [

18

O]water target to yield [

18

F]fluoride was followed by azeotropic drying of the Kryptofix [2.2.2]®/potassium carbonate/[

18

F]fluoride mixture. The reactive [

18

F]fluoride was then reacted with methyl iodide in anhydrous acetonitrile to yield methyl [

18

F]fluoride, which was isolated by gas chro- matography and trapped at liquid nitrogen tempera- tures. The purified methyl [

18

F]fluoride was then passed through a discharge chamber operating at 20–30 kV and 280 μA, and small amounts of F

2

(150 nmoles) and neon carrier gases were added to yield [

18

F]F

2

(7.5 GBq [

18

F]F

2

from 37 GBq [

18

F]fluoride). This [

18

F]fluoride-to-[

18

F]fluorine conversion process offers the option of obtaining high- specific-activity [

18

F]F

2

where the use of standard electrophilic [

18

F]fluorine targetry would lead to un- acceptably low specific activity products and/or nucle- ophilic radiolabeling methods utilizing [

18

F]fluoride are not practical.

Table 99.2. Cyclotron manufacturers and some currently available products Company Model Maximum Energy of Self-Shielded

Protons(p) and Deuterons (d)

CTI RDS 111 11 MeV (p) Yes

EBCO TR19 13-19 MeV (p) Option

9 MeV (d) optional

GE PETtrace 16.5 MeV (p) Option

8.4 MeV (d)

GE MINItrace 9.6 MeV (p) Yes

IBA Cyclone 10/5 10 MeV (p) No

5 MeV (d)

IBA Cyclone 18/5 18 MeV (p) No

5 MeV (d)

(4)

acid and treated with chromic acid. Radioactive bromine was distilled into an ammonia solution using a stream of nitrogen, and the resultant ammonium [

76

Br]bromide was dried and used as a source of [

76

Br]bromide for subsequent reactions (either nucleophilic or electrophilic following oxidation).

Another method of bromine-76 production utilized [

76

Se]selenium-enriched (96%) Cu

2

Se as the target ma- terial [28]. Irradiation with 17 MeV protons produced [

76

Br]bromine, which was separated from the solid target by thermal diffusion. Lower energy, 11 MeV cyclotrons have also been utilized to produce [

76

Br]bromide, but to date the yields have been low [29]. Bromine-76 has a more complex decay scheme than fluorine-18, and only about 57% of its transitions result in positron emission (Table 9.1). The positron emitted from

76

Br has a considerably higher kinetic energy than that from

18

F, resulting in higher patient dose per positron emission and lower imaging resolution.

Production of 124 I

Iodine-124 has been produced by the irradiation of 96% enriched [

124

Te]tellurium (IV) oxide with 15 MeV deuterons via the

124

Te(d,2n)

124

I nuclear reaction [30–32]. Iodine-124 has also been produced by the irra- diation of enriched [

124

Te]tellurium (IV) oxide with 15 MeV protons via the

124

Te(p,n)

124

I nuclear reaction [33]. Some work has been performed utilizing lower energy 11 MeV proton cyclotrons to produce [

124

I]iodide [29,34,35], but the yields have been rela- tively low. While

124

I has a relatively long half-life (4.2 days), there are problems associated with its use. Like

76

Br,

124

I has a complex decay scheme with only about 25% of its transitions resulting in positron emission, and the emitted positron has a relatively high energy compared to positrons from

18

F (Table 9.1). In addi- tion, several high-energy gamma rays of nuclear origin are emitted along with the positron. Despite these com- plications,

124

I has been used successfully to label PET radiopharmaceuticals because its long half-life pro- vides advantages over

18

F for imaging slow pharmaco- kinetic processes in vivo. In addition, it is possible to achieve relatively high-specific-activity products using electrophilic radiolabeling methods with

124

I.

The majority of PET radiohalogenations reported in the literature are nucleophilic fluorinations utilizing [

18

F]fluoride. The reasons for this include the availabil- ity of high amounts of [

18

F]fluoride from low- and medium-energy proton-only cyclotrons utilizing [

18

O]water target material and the generally higher specific activities achievable using [

18

F]fluoride. A variety of chemical reaction types are amenable to ra- diolabeling using [

18

F]fluoride. These can be divided into two principal catagories: SN2-type (substitution nucleophilic bimolecular) reactions with substrates containing leaving groups such as halides or alkyl sul- fonate esters, and aromatic nucleophilic substitution reactions utilizing activated aromatic systems with leaving groups such as nitro or trimethylammonium.

Radiofluorination via SN2 Reactions

In SN2-type reactions [6], nucleophiles attack the sub- strates at 180

º

opposite to the leaving groups resulting in configurational inversion at the carbon center fol- lowing substitution (Fig. 9.1). From a kinetic view- point, the reaction rates are largely determined by the structures of both the substrates and nucleophiles.

Substrates containing bulky substituents near the reac- tion center or poor leaving groups for substitution gen- erally decrease the reaction rate. Nucleophiles react with substrates as a function of their electron-donat- ing ability, and the fluoride anion is a poor nucleophile.

As a result, it has only been within the past 15 years that radiochemists have been able to successfully produce a wide variety of radiofluorinated compounds for PET imaging utilizing this type of reaction.

For SN2-type reactions, the best leaving groups are

the weakest bases, which is consistent with the princi-

ple that independently stable species make better

leaving groups. Of the halides, iodide is the best leaving

group and fluoride the worst. The conversion of an

alcohol to a sulfonic ester is a useful way to generate a

good leaving group. The triflate, tosylate, brosylate, no-

sylate, and mesylate groups are all better leaving

groups than halides. Leaving groups that have been uti-

lized the most for nucleophilic radiofluorination reac-

tions include triflate, tosylate, mesylate, bromide, and

iodide [1].

(5)

SN2-type radiofluorination reactions generally take place under basic or neutral conditions, as these condi- tions promote the presence of good nucleophiles (which are often strong bases). The effects of solvents on SN2-type reactions are variable and depend upon the charge dispersal of the reactants and subsequent transition state. An assortment of solvents have sup- ported high-yield radiofluorination reactions, and the solubility of the reactants appears to have played a larger role in solvent choice than their effects on reac- tion rates. The most common solvent choices (although by no means the only) are dipolar aprotic solvents in which both the alkali metal [

18

F]fluoride salts (for example, K[

18

F] or Cs[

18

F]) and the organic radiolabel- ing precursors generally show good solubility.

An important point in the general discussion of SN2- type radiofluorinations regards the propensity of fluoride to form tight ion pairs with metal cations.

Non-bound or non-coordinated nucleophiles are more reactive. Cryptands and polyaminoethers have been used to coordinate alkali metal cations, for example the potassium ion of K[

18

F]. This allows the [

18

F]fluoride anion to be less tightly paired with the cation (termed the “naked ion” effect) and subsequently more reactive [36]. Crown ethers, particularly 18-crown-6, have been used with K[

18

F] to increase solubility and promote the nucleophilicity of [

18

F]fluoride. Aminopolyethers (such as Kryptofix [2.2.2]®) have also been used with excel- lent results in a variety of aliphatic nucleophilic [

18

F]fluoride substitution reactions [37–39] Cesium and rubidium fluoride salts have been used as radiola- beling sources of reactive [

18

F]fluoride employing the concept that larger mono-valent cations bind less tightly to [

18

F]fluoride [40,41]. A variety of tetraalky- lammonium [

18

F]fluoride salts have been used widely in nucleophilic labeling reactions [42]. These salts are very soluble in a variety of organic solvents ranging from nonpolar to dipolar aprotic.

The use of alkyl halides as [

18

F]fluoride radiolabel- ing substrates has been viewed as attractive for a variety of compounds, particularly the simple di- halogen substituted alkanes. This approach has been used to produce

18

F-radiolabelled synthons such as 1- [

18

F]fluoro-3-bromopropane (Fig. 9.3), which was then used to radiolabel [

18

F]β-CFT-FP (radiochemical yield 2–3% decay-corrected to EOB) for use as a dopamine transporter radioligand [43]. Other synthons used in this manner include 1-[

18

F]fluoro-2-bromoethane and 1-[

18

F]fluoro-3-iodopropane [44–46].

Another example of the use of a halogen as the leaving group (Fig. 9.4) made use of an iodo group located adjacent to a carbonyl to incorporate [

18

F]fluoride into 21-[

18

F]fluoropregnenolone in a ra- diochemical yield of 20% [47].

Nucleophilic substitution with [

18

F]fluoride is also possible using benzylic halides as starting materials (Fig. 9.5), as evidenced by the synthesis of 3β-(4-[

18

F]fluoromethylphenyl)- and 3β-(2-[

18

F]fluoro- methylphenyl) tropane-2 β-carboxylic acid methyl esters for use as a dopamine transporter system radio- ligand [48]. The phenyltropane analog was obtained in 22% radiochemical yield (decay corrected to EOB), with chemical and radiochemical purities >99% of specific activities ranging from 2–5 Ci/ μmol.

In those cases where an alkyl alcohol is available, subsequent activation by the formation of the corre- sponding alkyl sulfonate ester derivative (tosylate, mesylate, triflate, 1,2-cyclic sulfate) has proven to be extremely useful in the preparation of alkyl [

18

F]fluorides. This reaction is general in scope and has been used to synthesize a variety of complex radio- ligands containing primary and secondary [

18

F]fluorides. In a manner analogous to that of the di- substituted haloalkanes, the displacement of mesylate [49,50], tosylate [51], and triflate groups [52] led to the synthesis of substituted ω-[

18

F]fluoroalkyl synthons.

Figure 99.3.

(6)

For example, the reaction of 3-bromopropyl triflate with [

18

F]fluoride produced 1-[

18

F]fluoro-3-bromo- propane [53].

The synthesis of 1-amino-3-[

18

F]fluorocyclobutane- 1-carboxylic acid provides an example of a nucle- ophilic substitution reaction with [

18

F]fluoride utilizing a triflate derivative, prepared from the corre- sponding alcohol, as the radiofluorination substrate [54]. This unnatural, non-metabolized amino acid (Fig. 9.6) was used to visualize malignant tumors and was produced in 12% radiochemical yield with a specific activity >1.5 Ci/μmol (>55 GBq/μmol).

The high-affinity dopamine D

2

receptor antagonist, [

18

F]fallypride (Fig. 9.7), was synthesized by a nucle- ophilic [

18

F]fluoride substitution reaction on the cor- responding tosylate in about 20% radiochemical yield at EOS [55].

Another example of the use of the tosylate leaving group to incorporate [

18

F]fluoride was the synthesis of 9-(4-[

18

F]fluoro-3-hydroxy methylbutyl)guanine ([

18

F]FHBG) [56]. [

18

F]FHBG (Fig. 9.8) was developed as a potential PET imaging agent to assess gene therapy. The masking of other reaction sites on the precursor molecule using trityl protection illustrates the ability to incorporate protecting group methodol- ogy into radiosynthetic strategies as a result of both the relatively long half-life of

18

F and the chemical sta- bility of the alkyl [

18

F]fluorides (see also the radiosyn- thesis of FDG). [

18

F]FHBG was prepared in 8–22%

radiochemical yield (decay corrected to EOB) with specific activities > 450 mCi/ μmol (16.7 GBq/μmol).

In some instances, the activated leaving group used to incorporate [

18

F]fluoride can also act as a protecting group for other functionalities present in the substrate.

Figure 99.4.

Figure 99.5.

Figure 99.6.

Figure 99.7.

(7)

This was the case for 3-O-methoxymethyl-16β,17β-O- epiestriol cyclic sulfone (Fig. 9.9), used as the precursor for the synthesis of 16 α-[

18

F]fluoroestradiol [57]. In this example, the cyclic sulfone acted as a protecting group for the 17β-hydroxyl functionality as well as ac- tivating nucleophilic displacement by [

18

F]fluoride at C-16. The axial methyl group at C-19 prevented attack from the β-face of the D-ring, and there was no evi- dence of displacement reactions at the C-17 position.

16 α-[

18

F]fluoroestradiol was prepared in 30–45%

radiochemical yield (decay corrected to EOB) with specific activities reported to be > 1 Ci/μmol (3.7 GBq/μmol).

Another example of a dual-mode leaving and protecting group can be found in the utilization of 2,3′-anhydro-5′-O-(4,4′-dimethoxytrityl)thymidine to produce 3′-deoxy-3′-[

18

F]fluorothymidine (FLT) (Fig.

9.10) for use as a cellular proliferation marker in a decay-corrected radiochemical yield of approximately

14% [58,59]. The 2,3′-anhydro structure not only acts as the leaving group for nucleophilic radiofluorination, but also serves as a protecting group for the 3-N- position of the pyrimidine ring.

The most frequently used PET radiopharmaceutical, 2-deoxy-2-[

18

F]fluoro-D-glucose (FDG), is currently produced utilizing [

18

F]fluoride-for-alkyl sulfonate ester radiolabeling methodology (Fig. 9.11). The in- crease in demand for FDG has led to significant effort directed towards the development of routine produc- tion methods as well as the design and construction of remote, automated systems dedicated to the synthesis of FDG.

FDG is presently synthesized using modifications of the method developed at the Julich PET Centre [37]. In the original method, aqueous [

18

F]fluoride was added to a solution consisting of Kryptofix [2.2.2]® and potassium carbonate dissolved in aqueous acetonitrile.

The residual water was removed by repeated azeotropic Figure 99.8.

Figure 99.9.

Figure 99.10.

(8)

distillations using anhydrous acetonitrile and a stream of nitrogen. The triflate precursor (1,3,4,6-tetra-O- acetyl-2-O-trifluoromethanesulfonyl- β-D-mannopyra- nose) was dissolved in acetonitrile and added to the dried [

18

F]fluoride. The reaction mixture was heated to reflux for five minutes. The resultant solution was passed through a C

18

Sep-Pak® cartridge. The residual aminopolyether was removed by washing the C

18

Sep- Pak® with 0.1 M hydrochloric acid. The radiolabeled acetylated carbohydrates were eluted into a second re- action vessel using tetrahydrofuran, and the ether was removed. Aqueous hydrochloric acid was added to the acetyl-protected intermediate (2-deoxy-2-[

18

F]fluoro- 1,3,4,6-tetra-O-acetyl- β-D-glucopyrranose), and the solution was heated at 130

º

C for 15 minutes. The product was purified by passage through an ion-retar- dation resin followed by an alumina column. The method was utilized as the basis of a computer-con- trolled automated synthesizer for the routine produc- tion of FDG [60]. Further modifications of the Julich methodology have led to the development of “one-pot”

syntheses for the production of FDG. These modifications include the substitution of tetramethy- lammonium carbonate for Kryptofix [2.2.2]®/potas- sium carbonate as the phase-transfer reagent and subsequent elimination of the C

18

Sep-Pak® cartridge- purification step. As a result of these modifications, the acidic hydrolysis was performed in the same reaction vessel. The reported radiochemical yield was 52% at the end-of-synthesis (EOS) with a total synthesis time of 48 minutes [61]. A similar “one-pot” modification was reported that retained Kryptofix [2.2.2]® as the phase-transfer reagent. Several Sep-Pak® cartridges were added to the system to remove unwanted Kryptofix [2.2.2]® and to prevent [

18

F]fluoride break- through. These modifications provided a radiochemi- cal yield of 65–70% decay-corrected to the end-of-bombardment (EOB) in a total synthesis time of approximately 50 minutes [62].

Toxicity concerns associated with Kryptofix [2.2.2]®

( LD

50

35 mg/kg in rats) have prompted the use of other phase-transfer agents, such as tetrabutylammonium hydroxide or tetrabutylammonium bicarbonate. This

modification has been incorporated into a commer- cially available synthesizer produced by Nuclear Interface, Inc. The Nuclear Interface module is flexible in that it can utilize either tetrabutylammonium bicar- bonate or Kryptofix [2.2.2]® as the phase-transfer reagent. In addition, the module can perform the hy- drolysis of the radiolabeled intermediate, 2-deoxy-2- [

18

F]fluoro-1,3,4,6-tetra-O-acetyl-β-D-glucopyrranose, under either acidic or basic (KOH) conditions. The module completes the radiosynthesis in less than thirty minutes with a reported radiochemical yield of ap- proximately 60% at EOS.

Another variation of the FDG radiolabeling scheme used an immobilized quaternary 4-aminopyridinium resin to isolate [

18

F]fluoride and subsequently incorpo- rate it into the

18

F-labelled intermediate [63,64]. The [

18

F]fluoride solution was passed across the resin column where [

18

F]fluoride was trapped, and the bulk of the enriched [

18

O]water was recovered downstream.

The resin-bound [

18

F]fluoride was dried by passing an- hydrous acetonitrile across the resin column while heating the column to approximately 100

º

C. A solution of the mannose triflate precursor in anhydrous ace- tonitrile was then passed over the heated resin column in either a slow single-pass or a reciprocating flow across the resin column. The solution containing the radiolabeled intermediate was then transferred to a hy- drolysis vessel where the acetonitrile was removed.

Following acid hydrolysis, FDG was purified in a manner analogous to the original method described above. The resin methodology formed the basis of a commercially available synthesis unit (PETtrace FDG MicroLab™, GE Medical Systems). This unit utilizes a disposable cassette system for the reaction column as well as disposable transfer and addition lines that facil- itate its set-up. Solid-phase support methodology that incorporates basic hydrolysis of the radiolabeled inter- mediate [65,66] has been implemented in the FDG syn- thesizer marketed by Coincidence Technologies, Inc.

The use of base decreased hydrolysis times to two

minutes at room temperature and resulted in no

epimerization. In addition, there are commercially

available pre-packaged reagent vials and pre-sterilized

Figure 99.11.

(9)

tubing systems for this automated synthesis module.

The module is equipped with a programmable logic controller that regulates the synthesis process. As a result, the system can be operated without a computer controller. The development of high-yield [

18

O]water targets for the production of [

18

F]fluoride, the capabil- ity of current generation cyclotrons to perform dual- target irradiations at relatively high beam currents, and the availability of efficient automated synthesis modules to produce FDG has made possible the pro- duction of multi-Curie (>74 GBq) amounts of FDG in a single cyclotron production run. This capability has significantly increased the utilization of FDG and has led to the growth of regional FDG production facilities that can supply a multitude of off-site users.

Radiofluorination via Aromatic Nucleophilic Substitution Reactions

While alkyl [

18

F]fluoride derivatives have seen frequent utilization in PET radiopharmaceuticals as noted above, significant effort also has been invested in radi- olabeling methods to incorporate [

18

F]fluoride into aromatic systems. These efforts include radiofluorina- tion methods to incorporate [

18

F]fluoride directly onto the aromatic ring as well as into prosthetic groups con- taining aromatic rings. Aromatic nucleophilic substitu- tions include:

(i) reactions in which the leaving group is activated by the presence of electron-withdrawing groups ortho and/or para to the leaving group;

(ii) reactions catalyzed by strong bases that proceed through an aryne (triple bond) intermediate; and (iii) reactions in which the nitrogen of a diazonium salt

is replaced by the nucleophile.

The first two examples can be classified as SNAr reac- tions, but the third example is an SN1-type reaction.

The first class of reaction is by far the most commonly used for aromatic radiofluorinations, wherein a leaving group is activated by the presence of ortho and/or para electron-withdrawing groups on the aromatic ring. An

approximate ranking of common substituents in order of decreasing activation ability includes:

NO

2

>CF

3

>CN>CHO>COR>COOR>COOH>Br>I>F>

Me>NMe

2

>OH>NH

2

[1,67]. The leaving group will also have an effect on the reaction rate. The following list is an approximate order of leaving group ability in aromatic nucleophilic substitution reactions:

NMe

3+

>NO

2

>CN>F>Cl,Br,I>OAr>OR>SR>NH

2

. There have been attempts to correlate the radiochemi- cal yields in nucleophilic radiofluorination reactions with the C-13 NMR chemical shifts of the correspond- ing fluoro-, nitro-, and trialkylammonium-substituted aryl aldehydes, ketones, and nitriles. While good agree- ment was found for the displacement of the substituted fluoro and nitro groups, the trialkylammonium group did not show the same correlation pattern [68,69].

The radiosynthesis of [

18

F]altanserin, a serotonin 5- HT

2A

receptor ligand, utilized the corresponding aro- matic nitro precursor in the nucleophilic substitution reaction with potassium [

18

F]fluoride in the presence of potassium carbonate and Kryptofix [2.2.2]® and il- lustrates activation of the nitro group by a carbonyl group situated para to the leaving group (Fig. 9.12).

The decay-corrected radiochemical yield was reported to be 20% [70].

As a result of the relatively long half-life of

18

F, there are several examples of multi-step radiosynthetic path- ways where the radionuclide is incorporated very early in the process. An example of a multi-step radiosyn- thetic pathway is the no-carrier added synthesis of 6- [

18

F]fluoro-L-DOPA [71]. In this case, [

18

F]fluoride was used as the radiofluorinating agent in the preparation of 3,4-dimethoxy-2-[

18

F]fluorobenzaldehyde from the corresponding nitro-substituted compound (Fig. 9.13).

The resultant

18

F-labeled product was then reacted in a enantiomerically pure variant of the 2-phenyl-5-oxa- zolone procedure to yield

18

F-labeled α,β-didehydro derivatives. Following enatioselective reduction and deprotection, 6-[

18

F]fluoro-L-DOPA was isolated in 3%

decay-corrected radiochemical yield with an enan- tiomeric excess >90%.

There are many examples of the use of aryl trialky- lammonium salts as alternatives to nitro-substituted

Figure 99.12.

(10)

aryl precursors. Aryl trialkylammonium salts tend to be more reactive and require milder conditions for [

18

F]fluoride incorporation. An example of this type of S

N

Ar reaction is the radiosynthesis of 4-[

18

F]fluorobenzyl iodide (Fig. 9.14) [41]. 4-Trimethyl- ammoniumbenzaldehyde trifluoromethanesulfonate in aqueous dimethyl sulfoxide was reacted with Cs[

18

F].

The resultant substituted [

18

F]fluorobenzaldehyde was reduced to the benzyl alcohol followed by treatment with hydriodic acid to yield 4-[

18

F]fluorobenzyl iodide in approximately 25% yield (EOS). This prosthetic group is amenable to incorporation into a variety of radiopharmaceuticals, including (+)-N-(4-[

18

F]fluoro- benzyl)-2 β-propanoyl-3β-(4-chlorophenyl)tropane [72].

The synthesis of [

18

F]norchlorofluoroepibatidine provides an example of the use of [

18

F]fluoride with trialkylammonium salts as the leaving group from a heteroaromatic ring [73]. Using the tert-BOC-pro- tected epibatidine derivative with trimethylammonium iodide as the leaving group, the desired

18

F-labeled compound was prepared in 70% radiochemical yield (Fig. 9.15). Subsequent deprotection and N-methyla- tion afforded overall radiochemical yields of 45–55%

for [

18

F]N-methyl-norchlorofluoroepibatidine with a specific activity of 2–6 Ci/ μmol at EOS.

An

18

F-radiolabeled prosthetic group approach similar to the use of [

18

F]fluorobenzyl iodide has been used to label oligonucleotides [74]. In this case, the desired prosthetic group, N-(4-[

18

F]fluorobenzyl)-2- Figure 99.13.

Figure 99.14.

(11)

bromoacetamide, was produced in a three-step syn- thesis that began with the nucleophilic aromatic radiofluorination of trimethylammonium benzoni- trile triflate. The resultant 4-[

18

F]benzonitrile was reduced to provide the radiolabeled benzyl amine, which was allowed to react with bromoacetyl bromide to yield the desired prosthetic group (Fig. 9.16). The radiochemical yield was approximately 12% and the specific activity of the resultant labeled oligonu- cleotide was 1 Ci/μmol (3.7 GBq/μmol) at EOS.

Similar approaches have been reported by other groups utilizing N-succinimidyl-4-[

18

F]fluoroben- zoate [75] as well as a solid-phase based approach using 4-[

18

F]fluorobenzoic acid [76].

Other Nucleophilic [ 18 F]Fluorination Reactions

A classic method for the synthesis of aromatic fluorides, known as the Balz–Schiemann reaction, in- volves the thermal decomposition of aryl diazonium

tetrafluoroborate salts (Fig. 9.17). While this method- ology has been applied to the production of aryl [

18

F]fluorides, it suffers several drawbacks [1]. The use of [

18

F]BF

4

as the counter anion results in low radio- chemical yields as well as low specific activity products.

There has been little utilization of this methodology for the synthesis of more complex radiofluorinated compounds. It should be noted that this reaction could allow the incorporation of [

18

F]fluoride into an aro- matic ring that is not activated to nucleophilic substi- tution reactions.

Figure 99.16.

Figure 99.15.

Figure 99.17.

(12)

Electrophilic Reactions with 18 F + (Low Specific Activity)

Fluorine is the most electronegative of all the elements.

Fluorine exists as a colorless to pale yellow corrosive gas (F

2

) that reacts with many organic and inorganic substances. Fluorine is a powerful oxidizing agent and attacks both quartz and glass, making its handling problematic. The use of fluorine gas as a carrier in the production of [

18

F]F

2

leads to several orders of magni- tude lower-specific-activity reaction products com- pared to methods using no-carrier added [

18

F]fluoride obtained from [

18

O]water targets.

Early synthetic methods for the production of FDG were based on electrophilic radiofluorination chem- istry (Fig. 9.18). The reaction of [

18

F]F

2

with 3,4,6-tri- O-acetyl glucal in fluorotrichloromethane (Freon-11) was the first synthetic method employed for this im- portant PET radiopharmaceutical [77,78]. One disad- vantage of this method, due to the highly reactive nature of F

2

, was the production of the protected fluoromannopyrranosyl leading to the undesired 2- deoxy-2-[

18

F]fluoro-D-mannose (FDM) derivative (Fig.

9.18). The use of an alternative fluorinating agent, acetyl [

18

F]hypofluorite, was proposed as a method for the routine production of FDG [79,80]. The use of

acetyl [

18

F]hypofluorite resulted in the regioselective (95%) synthesis of the desired glucose configuration under optimal reaction conditions. However, non- optimal conditions led to the production of undesired FDM in high yield [81]. In addition to this problem, electrophilic methods using [

18

F]F

2

to produce mono- fluorinated products also suffer from the loss of 50%

of the radioactivity. The development of high-yield [

18

F]fluoride targets and stereospecific nucleophilic radiofluorination chemistries have replaced elec- trophilic methods for the production of FDG.

One example of a direct electrophilic radiofluorina- tion of an activated aromatic substrate is the synthesis of a series of purine derivatives that have shown promise for PET imaging of a reporter system to assess viral gene therapy [82]. Earlier work demonstrated the capability of direct incorporation of fluorine into the C-8 position of a series of substituted purine deriva- tives [83,84]. The analogous radiofluorinations (Fig.

9.19) using [

18

F]F

2

led to the production of 8- [

18

F]fluoroganciclovir, 8-[

18

F]fluoropenciclovir, 8- [

18

F]fluoroacyclovir, and 8-[

18

F]fluoroguanosine. While this method does not require the use of protecting groups, the radiochemical yields were low (0.9–1.2%

decay-corrected to EOB). Nevertheless, sufficient mate- rial was produced to allow for the utilization of these [

18

F]fluoropurine derivatives in animal studies [85].

Figure 99.19.

Figure 99.18.

(13)

The direct electrophilic fluorination of aromatic rings has been accomplished using a variety of

18

F-radiolabeled electrophilic fluorinating agents including [

18

F]F

2

, xenon [

18

F]difluoride, and acetyl [

18

F]hypofluorite. These types of radiofluorinations are not generally regioselective. As an example, the reac- tion of 3,4-dihydroxyphenylalanine with [

18

F]F

2

using liquid hydrogen fluoride as a solvent (Fig. 9.20) yielded 2-, 5-, and 6-[

18

F]fluoro-L-DOPA in the ratio of 35:5:59, respectively [86].

The lack of regiospecificity in direct electrophilic radiofluorination reactions has resulted in the in- creased use of demetallation reactions. Regioselective demetallations have been used to great advantage with [

18

F]F

2

. Examples can be found in the literature for the use of aryl tin, mercury, silicon, selenium, and germa- nium compounds as substrates for radiofluorinations using [

18

F]F

2

. This type of reaction has been utilized to produce 6-[

18

F]fluoro-L-DOPA [87,88]. The most common precursor is an aryl substituted trialkyl tin derivative (Fig. 9.21). The decay-corrected radiochemi- cal yield was reported to be 29–37% using a protected trialkyl tin derivative with trifluorochloromethane as the reaction solvent, followed by acidic removal of the

phenol and amino acid protecting groups. This methodology has been utilized for the automated pro- duction of 6-[

18

F]fluoro-L-DOPA using a commercially available computer-controlled synthesis apparatus (Nuclear Interface, Inc.).

Another example of electrophilic regioselective fluorodemetallation is the radiosynthesis of [

18

F]WIN 35,428 ([

18

F] β-CFT) [89]. In this case, [

18

F] β-CFT was prepared by the electrophilic radiofluorination of 2 β- carbomethoxy-3β-(4-trimethylstannylphenyl)tropane using acetyl [

18

F]hypofluorite as the fluorination agent (Fig. 9.22) with a specific activity of 18–25 GBq/mmol and a yield of 0.9–2.0%.

In a manner analogous to the other halogens, fluorine will add to double bonds (Fig. 9.23). The ra- diosynthesis of [

18

F]2-(2-nitro-1[H]-imidazol-1-yl)-N- (2,2,3,3,3-pentafluoropropyl)-acetamide ([

18

F]EF5), a radiotracer used to assess tissue hypoxia, took advan- tage of this reactivity by using [

18

F]F

2

in trifluoroacetic acid to radiofluorinate the perfluoro alkene in 10–15%

radiochemical yield [90].

The synthesis of 5-[

18

F]fluoro-2′-deoxyuridine (Fig.

9.24) is another example of the addition of fluorine to double bonds. In this case, the formal addition of Figure 99.20.

Figure 99.22.

Figure 99.21.

(14)

“fluorine acetate” to the 5,6-double bond led to an in- termediate, which upon base catalyzed elimination of acetic acid yielded the target molecule in a radiochem- ical yield of 15–25% [91].

Electrophilic fluorination of carbanions using fluorinating agents such as perchloryl[

18

F]fluoride, N-[

18

F]fluoro-N-alkylsulfonamides, N-[

18

F]fluoropyri- dinium triflate has seen some utilization. A series of N-[

18

F]fluoro-N-alkyl sulfonamides have been synthesized using [

18

F]F

2

and shown to be suitable for use in radiolabeling a variety of structurally simple aryl lithium and aryl Grignard reagents [92].

However, the methodology has not been widely utilized to radiolabel more complicated target compounds.

Radiobromine and Radioiodine for PET

Fluorine-18 is not the only radiohalogen that has shown utility in the synthesis of PET radiopharmaceu- ticals. While there are no useful positron-emitting ra- dionuclides of chlorine or astatine, there are useful

positron-emitting radionuclides of both bromine (

76

Br) and iodine (

124

I). These radionuclides have suitable half-lives (

76

Br t

1–2

= 16.1 hours and

124

I t

1–2

= 4.2 days) for use in PET studies and can be produced in sufficient quantities to foster the development of a variety of radiopharmaceuticals.

Over the past 40 years, a great deal of radiochemistry effort has been devoted to methods for attaching single photon- and beta-emitting radioiodines, such as

125

I,

131

I, and

123

I, onto large and small molecules for in vitro and in vivo experimental uses [94–98]. These radiola- beling methods are also applicable to positron-emit- ting radioiodines, such as

124

I, and to a large extent to radiobromines as well [99–102].

As with most halogen chemistry, [

76

Br]bromide can be used either as a source of nucleophilic bromide or as a source of electrophilic bromine upon oxidation.

[

76

Br] β-CBT, a dopamine transporter radioligand (Fig.

9.25), has been radiolabeled using both nucleophilic and electrophilic radiobromination chemistry [103].

A variety of dopamine receptor ligands have been labeled using bromine-76 including [

76

Br]FLB 457, [

76

Br]FLB 463, [

76

Br]bromolisuride, [

76

Br]bromospiper- Figure 99.23.

Figure 99.24.

(15)

one, and [

76

Br]PE2Br [104–107]. The radiolabeling of [

76

Br]PE2Br, (E)-N-(3-bromoprop-2-enyl)-2β-car- bomethoxy-3 β-(4’-tolyl)nortropane illustrates the ca- pability of incorporating the radiobromine and radioiodine into vinylic positions (Fig. 9.26). This moiety offers increased stability for the halides com- pared to alkyl-substituted analogs. [

76

Br]PE2Br was synthesized in a radiochemical yield of approximately 80% using NH

4

[

76

Br] and peracetic acid with the vinyl tri-n-butylstannane substituted tropane analog.

The serotonin transporter ligand 5-[

76

Br]bromo-6- nitoquipazine also has been synthesized (Fig. 9.27) [108], as well as a norepinephrine transporter agent [

76

Br]MBBG [109,110].

The electrophilic radiobromination of metaraminol (Fig. 9.28) yielded a mixture of the 4- and 6-substituted bromometaraminols (17% and 38% non-decay cor- rected radiochemical yields, respectively), which were separable by HPLC. These compounds have shown promise as radiotracers for the myocardial norepi- nephrine reuptake system [111].

Bromine-76 has also been utilized to radiolabel intact monoclonal antibodies where its longer half-life allows for longer clearance times [112,113]. The syn- thesis of a radiobrominated thymidine analog ([

76

Br]FBAU) for use as a cellular proliferation marker for PET has also been reported [114]. Ammonium [

76

Br]bromide was used in an electrophilic destannyla- Figure 99.25.

Figure 99.26.

Figure 99.27.

(16)

tion reaction to prepare the 3′,5′-dibenzoyl protected analog, which yielded the desired [

76

Br]FBAU follow- ing base hydrolysis (Fig. 9.29).

Iodine-124 has been used to radiolabel intact mono- clonal antibodies, where its longer half-life allows for longer metabolic clearance times [115–117]. In a manner analogous to the radiobromination chemistry discussed above, iodine-124 also has been used in both nucleophilic and electrophilic radiolabeling reactions.

An example of this was the radiosynthesis of [

124

I] β- CIT (Fig. 9.30), which was labeled using both electrophilic iododestannylation and nucleophilic substitution via iodo-for-bromo exchange [118].

In addition, insulin has been labeled with

124

I on the fourteenth amino acid residue (tyrosine) using electro- philic radiolabeling conditions [119]. The nucleoside analog 2′-fluoro-2′-deoxy-1β-D-arabinofuranosyl-5- iodouracil (FIAU) has also been labeled using Na[

124

I]

and a stannylated uracil derivative as the precursor (Fig. 9.31) [120–122]. This radioiodinated derivative has been reported to result in significantly higher

specific accumulation of radioactivity compared to [

18

F]FHPG in tumor-bearing BALB/c mice [123].

Conclusions

Fluorine-18 is currently the most utilized PET radio- halogen as a result of its relatively facile production in large quantities, its convenient half-life, and its nearly optimal decay properties. Efficient incorporation of the

18

F-radiolabel into a variety of radiopharmaceuticals is possible using either nucleophilic routes with high specific activity [

18

F]fluoride or electrophilic routes with lower specific activity [

18

F]fluorine. The longer half-lives of

76

Br and

124

I can provide advantages over

18

F to image slower physiological processes, but the production of these radiohalogens is more demanding and their decay properties are more complex than those of

18

F. Thus,

18

F has become the radiohalogen of choice for a variety of PET imaging applications.

Figure 99.29.

Figure 99.28.

(17)

References

1. Kilbourn MR. Fluorine-18 labeling of radiopharmaceuticals, Nuclear Science Series NAS-NS-3203, Washington DC: National Academy Press,1990.

2. Stocklin G. Molecules labeled with positron emitting halogens.

Int J Rad Appl Instrum B 1986; 13:109–18.

3. Moerlein SM, Laufer P, Stocklin G, Pawlik G, Wienhard K, Heiss WD. Evaluation of

75

Br-labeled butyrophenone neuroleptics for imaging cerebral dopaminergic receptor areas using positron emission tomography. Eur J Nucl Med 1986; 12:211–16.

4. Mathis CA, Sargent III T, Shulgin AT. Iodine-122-labeled amphetamine derivative with potential for PET brain blood-flow studies J Nucl Med 1985; 26:1295–301.

5. Mathis CA, Lagunas-Solar MC, Sargent III T, Yano Y, Vuletich A, Harris LJ. A

122

Xe-

122

I generator for remote radio-iodinations.

Appl Radiat Isot 1986; 37:258–60.

6. March J. Advanced organic chemistry reactions, mechanisms, and structure, 4

th

edn. New York: John Wiley & Sons, 1992.

7. Fowler JS, Wolf AP. Positron emitter-labeled compounds: priori- ties and problems. In: Phelps ME, Mazziotta JC, Schelbert HR (eds). Positron emission tomography and autoradiography prin- ciples and applications for the brain and heart. New York: Raven Press, 1986, p 391–450.

8. McCarthy TJ, Welch MJ. The state of positron emitting radionu- clide production in 1997. Sem Nucl Med 1998; 28:235–46.

9. Welch MJ, Redvanly CS (eds). Handbook of radiopharmaceuti- cals: radiochemistry and applications. Sussex: John Wiley & Sons, 2002.

10. Kilbourn MR, Hood JT, Welch MJ. A simple

18

O water target for

18

F production. Appl Radiat Isot 1984; 35:599–602.

11. Kilbourn MR, Jerabek PA, Welch MJ. An improved [

18

O]water target for [

18

F]fluoride production. Int J Appl Radiat Isot 1985;

36:327–8.

12. Wieland BW, Hendry GO, Schmidt DG, Bida G, Ruth TJ. Efficient small-volume

18

O-water targets for producing

18

F-fluoride with low energy protons. J Label Compds Radiopharm 1986;

23:1205–7.

13. Ruth TJ, Buckle KR, Chun KS, Hurtado ET, Jivan S, Zeisler S.

A proof of principle for targetry to produce ultra high quantities of

18

F-fluoride. Appl Radiat Isot 2001; 55:457–61.

14. Schlyer DJ, Bastos M, Wolf AP. A quantitative separation of fluorine-18 fluoride from oxygen-18 water. J Nucl Med 1987;28:764.

15. Schlyer DJ, Bastos MA, Wolf AP. Separation of [

18

F]fluoride from [

18

O]water using anion exchange resin. Int J Rad Appl Instrument [A] 1990;41:531–3.

16. Jewett DM, Toorongian SA, Mulholland GK, Watkins GL, Kilbourn MR. Multiphase extraction: rapid phase-transfer of [

18

F]fluoride ion for nucleophilic radiolabeling reactions. Appl Radiat Isot 1988;39:1109–11.

17. Chan PKH, Firnau G, Garnett ES. An improved method for the production of fluorine-18 in a reactor. Radiochem Radioanal Lett 1974;19:237–42.

Figure 99.30.

Figure 99.31.

(18)

1983;34:1627–8.

22. Casella V, Ido T, Wolf AP, Fowler JS, MacGregor RR, Ruth TJ.

Anhydrous F-18 labeled elemental fluorine for radiopharmaceuti- cal preparation. J Nucl Med 1980;21:750–7.

23. Chirakal R, Adams RM, Firnau G, Schrobilegen GJ, Coates G, Garnett ES. Electrophilic

18

F from a Siemens 11 MeV proton-only cyclotron Nucl Med Biol 1995;22:111–6.

24. Nickles RJ, Daube ME, Ruth TJ. An O

2

target for the production of [

18

F]F

2

. Appl Radiat Isot 1984;35:117–22.

25. Bergman J, Solin O. Fluorine-18-labeled fluorine gas for synthesis of tracer molecules. Nucl Med Biol 1997;24:677–83.

26. Maziere B, Loc’h C. Radiopharmaceuticals labelled with bromine isotopes Appl Radiat Isot 1986;37:703–13.

27. Qaim SM. Recent developments in the production of

18

F,

75,76,77

Br, and

123

I Appl Radiat Isotop 1986;37:803–10.

28. Tolmachev V, Lövqvist A, Einarsson L, Schultz J, Lundqvist H.

Production of

76

Br by a low-energy cyclotron. Appl Radiat Isot 1998;49:1537–40.

29. Nickles RJ. Production of a broad range of radionucldies with an 11 MeV proton cyclotron. J Label Compd Radiopharm 1991;30:120–21.

30. Knust EJ, Dutschka K, Weinreich R. Preparation of

124

I solutions after thermodistillation of irradiated

124

TeO

2

targets. Appl Radiat Isot 2000;52:181–4.

31. Lambrecht RM, Sajjad M, Syed RH, Meyer W. Target preparation and recovery of enriched isotopes for medical radionuclide production. Nucl Instr Meth Phys Res 1989;282:296–300.

32. Weinreich R, Knust EJ. Quality assurance of iodine-124 produced via the nuclear reaction

124

Te(d,2n)

124

I. J Radioanal Nucl Chem Lett 1986;213:253–61.

33. Sheh Y, Koziorowski J, Balatoni J, Lom C, Dahl JR, Finn RD. Low energy cyclotron production and chemical separation of “no carrier added” iodine-124 from a reusable, enriched tellurium- 124 dioxide/aluminum oxide solid target. Radiochimica Acta 2000;88:169–73.

34. Qaim SM, Blessing G, Tarkanyi F, Lavi N, Bräutigam W, Scholten B et al. In: Cornell JC (ed). Proceedings of the 14

th

International Conference on Cyclotrons and their Applications, World Scientific, Singapore, p 541,1996.

35. Qaim SM, Hohn A, Nortier FM, Blessing G, Schroeder IW, Scholten B et al. (eds). Proceedings of the Eighth International Workshop on Targetry and Targetry Chemistry , St. Louis, USA, p 131, 2000.

36. Guibe F, Bram G. Réactivité SN

2

des formes dissociée et associée aux cations alcalins des nucléophiles anioniques. Bull Soc Chim Fr 1975;3:933–48.

37. Hamacher K, Coenen HH, Stocklin G. Efficient stereospecific syn- thesis of no-carrier-added 2-[

18

F]-fluoro-2-deoxy-D-glucose using aminopolyether-supported nucleophilic substitution.

J Nucl Med 1986;27:235–8.

38. Block D, Klatte B, Knochel A, Beckman R, Holm U. NCA [

18

F]-la- beling of aliphatic compounds in high yields via aminoether-sup- ported nucleophilic substitution. J Label Compd Radiopharm 1986;23:467–77.

39. Coenen HH, Klatte B, Knochel A, Schuller M, Stöcklin G.

Preparation of NCA [17-

18

F]fluoroheptadencanoic acid in high yields via aminopolyether-supported, nucleophilic fluorination.

J Label Compd Radiopharm 1986;23:455–66.

40. Shiue C-Y, Fowler JS, Wolf AP, Watanabe M, Arnett CD.

Syntheses and specific activity determinations of no-carrier- added (NCA) F-18-labeled butyrophenone neuroleptics-benperi- dol, haloperidol, spiroperidol, and pipamperone. J Nucl Med 1985;26:181–6.

41. Mach RH, Elder ST, Morton TE, Nowak PA, Evora PH, Scipko JG et al. The use of [

18

F]4-Fluorobenzyl iodide (FBI) in PET radio-

porter using positron emission tomography (PET). J Label Compd Radiopharm 2000;43:1235–44.

44. Block D, Coenen HH, Stocklin G. The NCA nucleophilic

18

F-fluorination of 1,N-disubstituted alkanes as fluoralkylation agents. J Label Compd Radiopharm 1987;24:1029–42.

45. Chi DY, Kilbourn MR, Katznellenbogen JA, Welch MJ. A rapid and efficient method for the fluoroalkylation of amines and amides. Development of a method suitable for the incorporation of the short-lived positron emitting radionuclide fluorine-18.

J Org Chem 1987;52:658–64.

46. Shiue C-Y, Bai L-Q, Teng R-R, Wolf AP. Syntheses of no-carrier- added (NCA) [

18

F]fluoroalkyl halides and their application in the syntheses of [

18

F]fluoroalkyl derivatives of neurotransmitter receptor active compounds. J Label Compd Radiopharm 1987;24:53–64.

47. Eng RR, Spitznagle LA, Trager WF. Preparation of radiolabeled pregnenolone analogs 21-fluoropregnenolone-21-

18

F, 21-fluoro- pregnenolone-3-acetate-21-

18

F, 21-fluoropregnenolone-7-

3

H, and 21-fluoropregnenolone-3-acetate-

3

H. J Label Compd Radiopharm 1983;20:63–72.

48. Petric A, Barrio JR, Namavari M, Huang S-C, Satyamurthy N.

Synthesis of 3β-(4-[

18

F]fluoromethylphenyl)- and 3β-(2- [

18

F]fluormethylphenyl)tropane 2 β-carboxylic acid methyl esters:

new ligands for mapping brain dopamine transporter with positron emission tomography. Nucl Med Biol 1999;26:529–35.

49. Kim SH, Jonson SD, Welch MJ, Katzenellenbogen JA. Fluorine- substituted ligands for the peroxisome proliferator-activated receptor gamma (PPAR?): potential imaging agents for metastatic tumors. J Label Compd Radiopharm 2001;44:S316.

50. de Groot T, Van Oosterwijck G, Verbruggen A, Bormans G.

[

18

F]Fluoroethyl β-D-glucoside and methyl β-D-3-[

18

F]fluoro-3- deoxyglucoside as ligands for the in vivo assessment of SGLT1 re- ceptors. J Label Compd Radiopharm 2001;44:S301.

51. Schirrmacher R, Hamkens W, Piel M, Schmitt U, Lüddens H, Hiemke C et al. Radiosynthesis of (±)-(2-((4-(2- [

18

F]fluoroethoxy)phenyl)bis(4-methoxy-phenyl)methoxy)ethyl- piperidine-3-carboxylic acid: a potential GAT-3 PET ligand to study GABAergic neuro-transmission in vivo. J Label Compd Radiopharm 2001;44:627–42.

52. Al-Qahtani MH, Hostetler ED, McCarthy TJ, Welch MR.

Improved labeling procedure of [

18

F]FFNP for in vivo imaging of progesterone receptors. J Label Compd Radiopharm 2001;44:S305.

53. Oh S-J, Choe YS, Chi DY, Kim SE, Choi Y, Lee KH et al. Re-evalu- ation of 3-bromopropyl triflate as the precursor in the prepara- tion of 3-[

18

F]fluoropropyl bromide. Appl Radiat Isot 1999;51:293–7.

54. Shoup TM, Goodman MM. Synthesis of [F-18]-1-amino-3-fluoro- cyclobutane-1-carboxylic acid (FACBC): a PET tracer for tumor delineation. J Label Compd Radiopharm 1999;42:215–25.

55. Mukherjee J, Yang Z-Y, Das MK, Brown T. Fluorinated benza- mide neuroleptics-III. Development of (S)-N-[(1-allyl-2-pyrro- lidinyl)methyl]-5-(3-[

18

F]fluoropropyl)-2,3-dimethoxybenzamide as an improved dopamine D-2 receptor tracer. Nucl Med Biol 1995;22:283–96.

56. Alauddin MM, Conti PS. Synthesis and preliminary evaluation of 9-(4-[

18

F]-fluoro-3-hydroxymethylbutyl)guanine ([

18

F]FHBG): a new potential imaging agent for viral infection and gene therapy using PET. Nucl Med Biol 1998;25:175–80.

57. Lim JL, Zheng L, Berridge MS, Tewson TJ. The use of 3-methoxymethyl-16 β,17β-epiestriol-O-cyclic sulfone as the precursor in the synthesis of F-18 16α-fluoroestradiol. Nucl Med Biol 1996;23:911–15.

58. Wodarski C, Eisenbarth J, Weber K, Henze M, Heberkorn U,

Eisenhut M. Synthesis of 3 ′-deoxy-3′-[

18

F]fluorothymidine with

Riferimenti

Documenti correlati

Through the ambivalence of her novel’s form, Smith appears also to imply that the postmodern self itself should learn to accept ambivalence as part of its existence, since

organelle outer membrane, outer membrane, positive regulation of cellular component organization, apoptotic mitochondrial changes, positive regulation of organelle organization,

In più, la ricchezza di appunti rivolti agli insegnanti ci informa come il sillabario non servisse solo al soldato apprendente; con particolare insistenza,

E questa mi sembra una situazione pericolosa, a cui bisogna stare molto attenti, bisogna vedere come tutto ciò ha funzionato in paesi periferici e come sta funzionando in

Several classes of real estate objects are possible from 1 to f (economy, economy-plus, average, business); s is the object status; sf is the number of statuses in accordance

Table 1 shows correlations between age, intent-based moral judgment separately for harm and help scenarios (i.e., mean moral score for scenarios with valenced intentions minus mean

Starting from the identified concepts (e.g. Information Source, Archaeo- logical Partition, Archaeological Unit), the archaeological data were modelled, inside the GeoUML Catalogue,

Figure 2 - 18 F FDG-PET/CT shows diffuse and symmetrical increased uptake in the fasciae of the upper and lower limbs, sparing both muscles and fat tis- sues on 18 F-FDG MIP