14
European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002 Abstract. Contrary to common belief, organometallic compounds exhibit remarkable stability in aerobic and even diluted aqueous solutions. Technetium-sestamibi (Cardiolite) is one of the most prominent examples of this class of compounds routinely used in nuclear medi- cine. This review summarises the recent progress in la- belling of biomolecules with organometallic complexes for diagnostic and therapeutic application in radiophar- macy and exemplifies in detail developments focussing on organometallic technetium- and rhenium-tricarbonyl technologies. The value of such technologies has been recognised and they have become a valuable alternative to common labelling methodologies. An increasing num- ber of groups have started to employ an organometallic precursor for the purpose of radioactive labelling of vari- ous classes of biomolecules, and the advantages and lim- itations of this new technique are compared with those of other labelling methods. The synthetic access to appro- priate precursors via double-ligand exchange or aqueous carbonyl kit preparation for routine application is de- scribed. Strategies and examples for the design of appro- priate bifunctional chelating agents for the Tc/Re-tricar- bonyl core are given. The functionalisation of biomole- cules such as tracers for the central nervous system (do- paminergic and serotonergic), tumour affine peptides (somatostatin receptors, neuroreceptors) and tumour binding single-chain antibody fragments is summarised. Where possible and appropriate, the in vitro and in vivo results in respect of these examples are compared with those obtained with classical 99m Tc/ 188 Re(V)- and 111 In- labelled analogues. The preclinical results show the in many ways superior characteristics of organometallic la- belling techniques. Keywords: Organometallic – Bioorganometallic – Tricar- bonyl – Radiopharmaceuticals – Biomolecule Abbreviations: MIBI, 2-Methoxyisobutylisonitrile 99m TcCO, [ 99m Tc(H 2 O) 3 (CO) 3 ] + – DAT, dopamine trans- porter – PgR, progesterone receptor – EgR, oestrogen re- ceptor – BFCA, bifunctional chelating agent – n.c.a., non-carrier-added – scFv, single-chain antibody frag- ments – mAb, monoclonal antibody – NET, norepineph- rine transporter – IPT, [N-(3-iodopropen-2-yl)-2β-carbo- methoxy-3β-(4-chlorophenyl) tropane] – β-CFT, 3β-(4- fluorophenyl)-tropane – Ac, acetyl – DTPA, diethylene triamine penta-acetate – IDA, iminodiacetate – BC, boranocarbonate – HYNIC, hydrazinonicotinic acid Eur J Nucl Med (2002) 29:1529–1542 DOI 10.1007/s00259-002-0900-8 Introduction Classical organometallic compounds, compounds with at least one direct transition metal-carbon bond, are extreme- ly rare in biological systems. The only naturally occurring organometallic compound so far discovered is the vitamin B 12 coenzyme, adenosyl cobalamine. Bio-organometallic science is a recently emerging discipline of potential im- portance for future directions in fields such as immunolo- gy, receptor research and assay development. In this fast- growing area of interdisciplinary research, numerous groups have recently reported remarkable progress. Upon being confronted with the term “organometal- lic”, even chemists will associate it with compounds which are air and moisture sensitive and insoluble in wa- ter. Contrary to common belief, organometallic com- pounds indeed exhibit remarkable stability in aerobic, aqueous solutions, offering a judicious choice of metals and ligand systems. As a matter of fact, an increasing number of compounds containing organometallic cores are under consideration for medical use. P. August Schubiger ( ) Department of Applied Bioscience, ETH Zurich, CH-8057 Zurich, Switzerland e-mail: [email protected] Tel.: +41-56-3102843, Fax: +41-56-3102849 Review article Current use and future potential of organometallic radiopharmaceuticals Roger Schibli 1 , P. August Schubiger 2 1 Center for Radiopharmaceutical Science ETH-PSI-USZ, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland 2 Department of Applied Bioscience, ETH Zurich, CH-8057 Zurich, Switzerland Published online: 6 August 2002 © Springer-Verlag 2002

Current use and future potential of organometallic radiopharmaceuticals

  • Upload
    ethz

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Abstract. Contrary to common belief, organometalliccompounds exhibit remarkable stability in aerobic andeven diluted aqueous solutions. Technetium-sestamibi(Cardiolite) is one of the most prominent examples ofthis class of compounds routinely used in nuclear medi-cine. This review summarises the recent progress in la-belling of biomolecules with organometallic complexesfor diagnostic and therapeutic application in radiophar-macy and exemplifies in detail developments focussingon organometallic technetium- and rhenium-tricarbonyltechnologies. The value of such technologies has beenrecognised and they have become a valuable alternativeto common labelling methodologies. An increasing num-ber of groups have started to employ an organometallicprecursor for the purpose of radioactive labelling of vari-ous classes of biomolecules, and the advantages and lim-itations of this new technique are compared with those ofother labelling methods. The synthetic access to appro-priate precursors via double-ligand exchange or aqueouscarbonyl kit preparation for routine application is de-scribed. Strategies and examples for the design of appro-priate bifunctional chelating agents for the Tc/Re-tricar-bonyl core are given. The functionalisation of biomole-cules such as tracers for the central nervous system (do-paminergic and serotonergic), tumour affine peptides(somatostatin receptors, neuroreceptors) and tumourbinding single-chain antibody fragments is summarised.Where possible and appropriate, the in vitro and in vivoresults in respect of these examples are compared withthose obtained with classical 99mTc/188Re(V)- and 111In-labelled analogues. The preclinical results show the inmany ways superior characteristics of organometallic la-belling techniques.

Keywords: Organometallic – Bioorganometallic – Tricar-bonyl – Radiopharmaceuticals – Biomolecule

Abbreviations: MIBI, 2-Methoxyisobutylisonitrile –99mTcCO, [99mTc(H2O)3(CO)3]+ – DAT, dopamine trans-porter – PgR, progesterone receptor – EgR, oestrogen re-ceptor – BFCA, bifunctional chelating agent – n.c.a.,non-carrier-added – scFv, single-chain antibody frag-ments – mAb, monoclonal antibody – NET, norepineph-rine transporter – IPT, [N-(3-iodopropen-2-yl)-2β-carbo-methoxy-3β-(4-chlorophenyl) tropane] – β-CFT, 3β-(4-fluorophenyl)-tropane – Ac, acetyl – DTPA, diethylenetriamine penta-acetate – IDA, iminodiacetate – BC,boranocarbonate – HYNIC, hydrazinonicotinic acid

Eur J Nucl Med (2002) 29:1529–1542DOI 10.1007/s00259-002-0900-8

Introduction

Classical organometallic compounds, compounds with atleast one direct transition metal-carbon bond, are extreme-ly rare in biological systems. The only naturally occurringorganometallic compound so far discovered is the vitaminB12 coenzyme, adenosyl cobalamine. Bio-organometallicscience is a recently emerging discipline of potential im-portance for future directions in fields such as immunolo-gy, receptor research and assay development. In this fast-growing area of interdisciplinary research, numerousgroups have recently reported remarkable progress.

Upon being confronted with the term “organometal-lic”, even chemists will associate it with compoundswhich are air and moisture sensitive and insoluble in wa-ter. Contrary to common belief, organometallic com-pounds indeed exhibit remarkable stability in aerobic,aqueous solutions, offering a judicious choice of metalsand ligand systems. As a matter of fact, an increasingnumber of compounds containing organometallic coresare under consideration for medical use.

P. August Schubiger (✉)Department of Applied Bioscience, ETH Zurich, CH-8057 Zurich, Switzerlande-mail: [email protected].: +41-56-3102843, Fax: +41-56-3102849

Review article

Current use and future potential of organometallic radiopharmaceuticalsRoger Schibli1, P. August Schubiger2

1 Center for Radiopharmaceutical Science ETH-PSI-USZ, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland2 Department of Applied Bioscience, ETH Zurich, CH-8057 Zurich, Switzerland

Published online: 6 August 2002© Springer-Verlag 2002

It is not only recently that nuclear medicine and ra-diopharmacy have played a pioneering and leading rolein the development of clinically applicable, organometal-lic drugs. One of the most widely used SPET imagingagents in routine nuclear medicine, technetium-99m ses-tamibi (Cardiolite, Fig 1), is a classical organometalliccompound. Originally developed as a myocardial perfu-sion agent [1], it is nowadays also successfully appliedfor tumour imaging and the detection of multidrug resis-tance [2, 3]. In fact, Cardiolite is the first example of anorganometallic complex routinely used in nuclear medi-cine.

Compared with the “first-generation” radiopharma-ceuticals, the prerequisites for a novel, e.g. target-specif-ic, radiopharmaceutical are greater with respect to its bi-ological activity. The compound has to maintain as muchaffinity and selectivity as possible for the selected targetbut not for other organs and tissue.

Since many of the interesting radionuclides and par-ticularly nuclides with potential therapeutic applicationare transition metals, there is a need for appropriate andefficient labelling procedures for various biomolecules.In this context, the “artificial” character of organometal-lic transition metal compounds might create distinct ad-vantages compared with “natural” drugs, which are oftensubject to unwanted rapid metabolism in vivo. Thus, or-ganometallic approaches might offer a valuable alterna-tive to common labelling protocols.

In the chemical literature there is no sharp definitionfor organometallic compounds. Thus, beside “classical”organometallics, transition metal complexes with or-gano-phosphorus, -sulphur or -selenium chelates canalso be regarded as organometallics. Applying thisbroader definition, many other, predominantly phospho-rus- and sulphur-based complexes developed in radio-pharmacy and used in nuclear medicine can be regardedas organometallics.

For the sake of consistency and clarity, this review ar-ticle will mainly focus on recent developments and theapplication of “classical” organometallic compounds of

technetium and rhenium. The productivity in this field inthe past decade merits an overview of the progress andfuture perspectives of this class of compounds in respectto radiopharmaceutical and nuclear medical applications.

Prerequisites for application of organometalliccompounds

The discovery of a new class of radiopharmaceuticalsstands at the beginning of a long development process.Several critical issues have to be addressed to satisfy theclinical requirements. The preparation of such com-pounds has to be simple, preferably in a kit formulation.Furthermore, the synthesis has to be safe (conditionswhich can be handled within a routine clinical environ-ment) and rapid (several minute to a few hours, also de-pending on the half-life of the radiometal), and shouldresult in products of high radiochemical purity (≥90%).Therefore, the principal limitations are the following: (a)any preparation has to be performed in physiological me-dia; (b) no purification of the final product should benecessary; (c) the reaction time should be short. Scrutinyof the chemical literature shows that these prerequisitesobviously made the introduction and use of organometal-lics rather unattractive until recently.

The technetium- and rhenium-tricarbonyl core

During the past few years the emphasis of technetium-and rhenium-based agents has gradually been shiftingfrom inorganic chemistry to biochemistry, focussing onthe nature of the biological group attached to the metalchelate. Many efforts have been undertaken to “hide” thetechnetium core and to adapt it to the characteristics ofthe corresponding biomolecule. It has been hypothesisedthat the smaller the technetium complex, the higher thelikelihood that the biological activity will not be altered.Stabilisation of the metal complex in vivo is another im-portant issue. It is well known that beside the thermody-namic stability of a complex, its kinetic stability or inert-ness is of equal, and sometime greater importance for anapplication in vivo.

It is unlikely that significant improvements in the la-belling of biomolecules with technetium and rheniumcan be expected with classical nitrogen/sulphur/phospho-rus-based ligand systems, because they have been op-timised during the past 20 years. Thus, further advancesare more likely when novel ligand systems, such as HYNIC or lower oxidation states, are explored.

One of the most promising and developed organome-tallic cores for labelling of biomolecules is the techne-tium- and rhenium-tricarbonyl core. The metal centre isin the low oxidation state +I and is therefore chemicallyvery inert. The M(CO)3 core (M = Tc, Re) is very com-pact, with an almost spherical shape. If the octahedral

1530

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 1. Structure of Tc-sestamibi (Cardiolite), the first clinicallyapplied organometallic compound in nuclear medicine

coordination sphere is “closed” with an appropriate li-gand system, the metal centre will be efficiently protect-ed against further ligand attack or re-oxidation. In con-trast, the “open” quadratic pyramidal structure of Tc(V)-oxo complexes with a tetradentate chelate is character-ised by unprotected sides, which are prone to ligand at-tack and protonation, which leads to decomposition ofthe original complex. The corresponding Re(V) com-plexes are even more affected by these characteristics.This has to date possibly been the greatest hindrance toextended therapeutic studies employing rhenium iso-topes. Furthermore, a qualitative comparison of the tri-carbonyl core with the widely employed complexes oftechnetium and rhenium in the oxidation state +V, suchas Tc(V)-MAG3, reveals a significantly reduced size(Fig. 2).

Schubiger’s group at PSI succeeded in developing anormal pressure preparation and a fully aqueous-basedpreparation of the precursor [M(H2O)3(CO)3]+ (M =99mTc, 99Tc, Re) in high yields and with excellent (ra-dio)chemical purity [4, 5]. The precursor can be obtainedby reduction and carbonylation of pertechnetate by inter-action with borohydrides and atmospheric pressure ofcarbon monoxide. This procedure circumvents the mostcommon starting materials for tricarbonyl compounds,decacarbonyl and halopentacarbonyl. Although the pub-lished preparation of [99mTc(H2O)3(CO)3]+, abbreviated99mTcCO, was suitable for research purposes, it still re-lied on toxic gaseous carbon monoxide. For a commer-cial radiopharmaceutical kit preparation this is the sub-ject of some concern. The use of solid, stable and non-toxic potassium boranocarbonate, K2[H3BCO2], whichreleases CO upon hydrolysis and can reduce Tc(VII) toTc(I) concomitantly, can be regarded as the ultimatebreakthrough permitting broader application of organo-metallic precursors [6]. The kit will soon be made avail-able for research purposes (Fig. 3). The recently optimi-sed preparation of the corresponding rhenium precursor

[188Re(H2O)3(CO)3]+, which promises to be useful forfuture therapeutic applications, differs only slightly interms of reducing agents and yields [7].

Closely related to the M(CO)3 subgroup is thecpM(CO)3 class of molecules, where cp stands for cyclo-

1531

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 2. Qualitative size comparison of the organometallic precur-sor [Tc(H2O)3(CO)3]+ (left) and Tc-MAG3 (right) based on X-rayanalyses. Purple = technetium, red = oxygen, grey = carbon, blue= nitrogen, yellow = sulphur. Hydrogen atoms are omitted

Fig. 3. First kit for the preparation of [99mTc(H2O)3(CO)3]+ start-ing from pertechnetate in saline [6]

Fig. 4. A Alberto’s method for preparation of organometallic tech-netium-cyclopentadienyl-tricarbonyl derivatives in aqueous mediastarting from pertechnetate [54]. B Wenzel’s method via double-li-gand-transfer reaction in organic media [9]

pentadienyl. The cpM(CO)3 core is highly lipophilic,making it particularly interesting for functionalisation ofsteroids and drugs that have to pass the blood-brain bar-rier. The fact that the cpM(CO)3 (M = Re) moiety can becoupled to biomolecules without affecting the bindingaffinities has been demonstrated [8]. The inherent advan-tage of the cp ligand is the low molecular weight and thestability of the resulting half-sandwich complex. Thefirst synthesis of functionalised cp derivatives and 99mTcwas reported by Wenzel (Fig. 4B) [9]. 99mTcO4

– was re-duced in presence of ferrocene derivatives andMn(CO)5Br. Katzenellenbogen and co-workers have re-cently presented a modification of this “double ligandtransfer” (DLT) reaction [10, 11]. However, the harsh re-action conditions in organic media and the often pooryields preclude practical application. An elegant, fullyaqueous-based preparation of cp99mTc(CO)3 derivativeswas recently published by Alberto and co-workers(Fig. 4A). The key step for the unique preparation in wa-ter is the introduction of electron-withdrawing substitu-ents, enabling deprotonation of the cp-ring at physiologi-cal pH. As a consequence, straightforward functionali-sation of biomolecules as well as labelling with the99mTc(CO)3 core under reasonable conditions and withsatisfactory yields could be achieved for the first time.

Ligand systems for the M(CO)3 core

The M(CO)3 core was not designed as a “stand-alone”radiopharmaceutical like, for example, Tc-sestamibi, butas a precursor for simple labelling procedures and easierand more appropriate functionalisation of biomolecules.Consequently, various bifunctional ligand and chelatingagents (BFCAs) have been designed and tested. Thethree carbonyl and the three vacant coordination sidesare facially arranged around the octahedral metal centre.This has a favourable influence on the size and geometryof the chelating units. The oxidation state +I allows theapplication of a broad variety of donor and acceptor at-oms [12].

In the search for appropriate mono-, bi- and tridentateligand systems, N-heterocycles such as imidazoles, pyri-dines and pyrazoles, amides, carboxylic acids, thio-ethers, thiols, phosphines and combinations thereof haveproven to coordinate efficiently to the tricarbonyl core(Fig. 5). Some of these functionalities also correspond toside chains of amino acids and are, therefore, of particu-lar interest for labelling of peptides and proteins. Egli etal. have investigated the ability of amino acids and ami-no acid fragments to react with the 99mTc-tricarbonylcore [13]. The most important finding was that histidinereacts quantitatively with the organometallic precursor atconcentrations as low as 10–6 M at 75°C. The extraordi-narily high capacity of imidazole to build and stabilise

1532

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 5. Examples of bidentateand tridentate chelating sys-tems for the functionalisationof biomolecules for labellingwith the technetium- and rheni-um-tricarbonyl core

hydrogen bonds is the major reason for this characteris-tic.

The low oxidation state of the technetium and rheni-um centre also allows the use of “exotic” ligand systemssuch as isonitriles (as in the case of, for instance, MIBI)or cyclopentadienes. Particularly the cps are of great im-portance, since they represent the one of the smallest“tridentate” chelates for the tricarbonyl core.

The high flexibility of the M(CO)3 core regarding ap-propriate ligands and consequently the simple function-alisation of biomolecules with mono- and bidentate che-lating systems is convincing and tempting from a chemi-cal point of view. However, pharmacokinetic experi-ments have unveiled some characteristics and limitationsregarding the minimal denticity of the chelate for opti-mal clearance rates and stability of model complexes invivo [14]. It has been observed that 99mTc-tricarbonylcomplexes which are coordinated with a tridentate che-lating system reveal good stability when challenged inhuman plasma and with excess cysteine, histidine or glu-tathione. These complexes have also shown very goodclearance from the blood pool and all tissue and organswhen tested in BALB/c mice (Fig. 6). In contrast, com-plexes which are only coordinated in a bidentate fashionshow significant aggregation with plasma proteins in vit-ro and in vivo. They are significantly retained in theblood and in the organs of excretion, such as the liver

1533

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 6. Biodistribution of two examples of tridentate and two ex-amples of bidentate coordinated 99mTc-tricarbonyl complexes inBALB/c mice, 24 h p.i.

Table 1. Selected examples of novel technetium/rhenium-tricarbonyl labelled compounds for potential radiopharmaceutical application

Biomolecule/ligand Target/potential application Ref.

1. Small molecules (<1,000 Da)

Biotin Avidinylated mAb/tumour pre-targeting [17]Glucose Tumours [18, 19]Oestrogen Oestrogen receptor/breast cancer [20, 21, 22, 23, 24, 25, 26]Progesterone Progesterone receptor/breast cancer [27]Tamoxifen Oestrogen receptors/breast cancer [20]WAY 100635 Serotonergic system [53]Piperidine derivatives Sigma receptors on tumours [28, 29, 30]Haloperidol Dopamine D2 receptors [9]Tropane Dopamine receptors [9, 31, 32]Benzazepine Dopamine D1 receptors [33]MIBI Multidrug resistance/renal imaging [34, 35]Etomidate Adrenal cortex/adrenal cortical tumours [36]

2. Medium size molecules (1,000<Da<15,000)

Somatostatin Somatostatin receptors [37, 38]Neurotensin Neurotensin receptors [39, 40, 41]Neuropeptide Y (NPY) NPY receptors [42]Bombesin Bombesin receptors/small-cell lung cancer [43]Bitisatin Glycoprotein IIb/IIIa receptor/thrombus imaging [44]

3. Large molecules (>15,000 Da)

ScFvs Colon cancer/bladder cancer [45, 46]Surfactant protein B Acute respiratory distress syndrome [47]MAb Bladder cancer [48]

and the kidneys. The overall charge and the lipophilicityof the complexes seem to play only a subordinate role inthese effects, to which several factors may contribute.Among these, the major one may be the fact that tricar-bonyl complexes with bidentate ligands still have a va-cant coordination side, occupied by a substitution labilewater molecule or chloride [15, 16]. Complexation offunctional groups of plasma proteins via this coordina-tion side can explain the prolonged retention in the bloodpool and (consequently) in all organs and tissue. Furtherinvestigations are necessary to confirm this assumptionand to reveal the exact mechanism in vivo. Therefore, al-though the synthetic complexity may be greater, it seemsmore favourable to functionalise biomolecules with tri-dentate chelates as this will yield a better pharmacoki-netic behaviour.

Radiopharmaceutical application of organometalliccompounds of technetium and rhenium

The number of potential applications of tricarbonyl tech-nology which are currently being exploited is remarkable(Table 1). The advantages of the tricarbonyl moiety arevaluable for the labelling of both small biomolecules(<1,000 Da) and large proteins (>15,000 Da).

Examples of small molecules labelled with Tc/Re-tricarbonyl

A particular challenge for the new generation of radio-pharmaceuticals is the labelling of small receptor-target-ing molecules with 99mTc. Especially neuroreceptor-tar-geting molecules have been the subject of extensive in-vestigation in recent years [49]. The feasibility and highpotential of such radiopharmaceuticals was demonstratedby 99mTc-TRODAT-1, a tropane analogue [50, 51]. Abalanced lipophilicity and a low molecular weight areprerequisites of such compounds. A crucial point for allreceptor-based radiopharmaceuticals is high specific ac-tivity, necessary in order to avoid receptor saturation andunwanted pharmacological side-effects.

These requirements are often hard to meet withTc(V) labelling techniques. Furthermore, most of thetetradentate ligand systems produce syn/anti-stereoiso-mers, which can differ significantly in their biologicalcharacteristics, particularly in the case of small mole-cules. The high labelling efficiency of the tricarbonylcore, the small overall size and the organometalliccharacter offer valuable means of overcoming existinghurdles. TROTEC-1 (Fig. 7A) represents the first ex-ample of a tropane derivative (β-CFT) with exception-ally high affinity in vitro to cloned human DAT, func-tionalised with a thioether/carbonyl moiety [32]. Func-tionalisation of the tropane ring was accomplished witha 4,7-dithiaoctaonic acid at position 2-β. While this

system does not overcome the problem of stereoisomerformation, it is promising because of the straightfor-ward functionalisation strategy via an ester group, thesmall size and the high lipophilicity of the complex.The rhenium complex is less polar than Tc(V)-oxo mol-ecules and revealed a tenfold enhanced affinity towardsmonoamine transporters compared with native β-CFTor "3+1” complexes derived from IPT [52]. IC50 valuesfor the analogue are 7.3±1.1 nM for NET (β-CFT,834±45 nM) and 71±1.4 nM for 5HTT (β-CFT,759±47 nM). The high affinity was explained by an en-hanced lipophilic interaction of the organometallic corewith the transporter.

In related efforts, Katzenellenbogen and co-workershave recently investigated the potential use of η5-cyclo-pentadienyltricarbonyl rhenium and technetium deriva-tives of tropane for DAT imaging [31]. Functionalisat-ion of the tropane analogues was accomplished via N-8-alkylation of nortropane or substitution at the paraposition of the 3β-phenyl group (Fig. 7A). The doubleligand transfer reaction was applied to insert the metal-tricarbonyl core. A strong dependence of the affinitiesfor all three monoamine transporters (DA, 5-HT andNE) on the position of functionalisation could be dem-onstrated. The N-8-substituted tropanes showed affini-ties in the low nanomolar range, whereas the 3β-substi-tuted systems had negligible affinities. These results

1534

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 7. Organometallic derivatives of tropane for the DAT (A) andligands for the serotonergic system (B) and their affinity for thetargeted receptor subtype

are in agreement with observations that the effect ofsubstituents at the para position of 3β-phenyl cannot bepredicted with our current knowledge and is not direct-ly related to the introduction of the organometallic la-bel.

Arylpiperazine is among the most thoroughly studiedCNS ligands for the 5-HT1A subclass of serotonergic re-ceptor. 1-(2-Methoxyphenyl)-piperazine, a truncated de-rivative of WAY 100635, has been functionalised withcyclopentadien and bidentate Schiff-base chelates, en-abling labelling with 99mTcCO with specific activities ofup to 30 GBq/µmol (Fig. 7B) [53]. The Schiff base com-plex revealed an IC50 value of 5±2 nM for the 5-HT1A re-ceptor. For 5-HT2A, dopamine D2 receptors and 5-HTand D transporters, the IC50 values were in the micromo-lar range. Thus, the biological affinity and selectivity ofthe native compound could be preserved in the organo-metallic derivatives. The complexes are stable in physio-logical phosphate buffer for at least 24 h at 37°C. For thepreparation of the cp-arylpiperazine derivative in yields>95%, a one-pot, single-step synthesis was described,starting directly from aqueous [99mTcO4]– [54]. This canbe regarded as significant progress towards routine ap-plication of the tricarbonyl technology, and of the cp-li-gand system in particular.

Metal carbonyl complexes of steroids have been thesubject of intensive investigation for some time. Jaouenand co-workers have exploited their use as a “cold” bio-logical probe for immunological assays, called CIMA(carbonyl metal immuno-assay) [55, 56, 57, 58]. Thisgroup and others have recognised at a very early stagethe potential of this class of compounds for the diagnosisand therapy of steroid receptor-positive breast cancer [8,9, 22, 23]. The groups of Johannsen and Katzenellenb-ogen have synthesised various 17β-progesterone and 7α-oestradiol dithioether and cyclopentadien complexes ofrhenium(I)-tricarbonyl (Fig. 8). The bioconjugates weretested in vitro for binding affinity towards PgR and EgR.The relative binding affinities (RBA; oestradiol =100%;

RU5020 =100%) of organometallic oestradiol deriva-tives were found to be only slightly lower (15%–23%RBA at 25°C) than those of “3+1” and “4+1” complexesof rhenium(III/V) (9%–45% RBA at 25°C) [59]. The dif-ferences in RBA can partially be explained by the higherlogPo/w values measured for the organometallic deriva-tives. RBA was also found to depend on the nature of thespacer between the metal chelate and the steroid moiety.Similar observations and tendencies have been reportedfor the progestin complexes [27]. For both examples, theorganometallic cyclopentadienyl-tricarbonyl systemswere superior to the dithioether-tricarbonyl systems interms of RBA for the corresponding receptors. Thismight be attributed to the diastereomeric nature of the di-thioether complexes and the presence of bulky bromineatoms in the metal coordination sphere. Synthesis of thecorresponding 99mTc analogues and biodistribution stud-ies will help to clarify the usefulness of these systems aseffective imaging agents for PR- and ER-positive breastcancer.

Examples of peptides labelled with Tc/Re-tricarbonyl

The most advanced branch where tricarbonyl technologyis extensively tested and applied is in combination withtumour affine peptides. Tricarbonyl technology can bebeneficial in several respects:

1. The labelling efficiency gives rise to high specific ac-tivities.

2. Purification of the labelled peptides is not usuallynecessary.

3. The convenient functionalisation strategy for peptides(applicable on a solid-phase synthesiser) is a furtheradvantage.

In general, small hydrophilic peptides distribute uni-formly and penetrate readily in tissue and clear efficient-ly from the circulation. Radiolabelling can result in im-portant changes in hydrophilicity and charge, with con-sequences for biodistribution and tissue kinetics. A “neu-tral” and “innocent” labelling method is therefore ofgreat importance.

Somatostatin analogues such as octreotide are amongthe most successful and thoroughly studied compoundsfor systemic radiotherapy and diagnosis. 111In-DTPA-D-Phe1-octreotide is clinically established and represents abenchmark for any further development of peptide-basedradiopharmaceuticals. Apart from HYNIC-functionalisedoctreotide analogues [60, 61], the 99mTc-99m labelling ofoctreotide has met with rather limited success to dateowing to the difficulties with disulphide bond reductionin the presence of stannous chloride in direct as well asindirect labelling approaches. Since Tc/Re-tricarbonyldoes not react with the disulphide bridges, the advantageof employing this technology is evident. 0Tyr3octreotate

1535

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 8. Organometallic derivatives of oestradiol (A) and progester-one (B) and the relative binding affinity compared with native oes-tradiol and RU5020

1536

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 9. Octreotate derivativesfunctionalised for labellingwith 99mTcCO. Potential metal-coordinating groups are la-belled in bold

Fig. 10. Gamma-camera images of A [99mTc(CO)3-Nα-Ac-His-0Tyr3octreotate]0, B [99mTc(CO)3-His-0Tyr3octreotate]+ and C [99mTc(CO)3-DTPA-0Tyr3octreotate]3– compared with D [111In-DTPA-0Tyr3octreotate]– in male Lewis rats bearing CA20948pancreatic tumours, 4 h p.i. B, Bladder; K, kidneys; L, liver; T,tumour

tivity in the cells was much higher in the case of themultiple anionic charged complexes (60.1%±4.6% reten-tion for [99mTc(CO)3-DTPA-0Tyr3octreotate]3– vs 17.9%±3.4% retention for [99mTc(CO)3-Nα-Ac-His-0Tyr3octreo-tate]0, after 16 h at 37°C). Biodistribution experimentswere performed in male Lewis rats bearing CA20948pancreatic tumours. Specific uptake could be detected inall somatostatin receptor-positive tissues. It could beshown that more negative charged BFCAs provide aslightly greater tumour retention and predominantly uri-nary excretion. Yet, [99mTc(CO)3-Nα-Ac-His-0Tyr3oct-reotate]0 showed excellent tumour-to-blood ratios of16:1 30 min p.i. and 33:1 4 h p.i (Table 2). These dataare comparable with those published for 99mTc-HYNIC-0Tyr3octreotate and other high-affinity somatostatin re-ceptor binding peptides such as 99mTc-P587 and 99mTc-P829 [60, 61, 62]. A tendency towards better in vivoproperties was observed if the peptide had been function-alised with multidentate or tridentate instead of histidine(bidentate chelate) only. This observation is a direct re-flection of the clearance characteristics of tridentate andbidentate coordinated 99mTcCO model complexes men-tioned earlier.

analogues functionalised with various BFCAs have beentested for labelling with 99mTcCO (Fig. 9) [38]. TheBFCAs gave rise to complexes of different overallcharge (+1 to –3). Aromatic BFCAs such as histidine(bidentate chelate) and Nα-Ac-His (tridentate chelate)gave rise to very high specific activities of 110 GBq/µmoland 220 GBq/µmol, respectively, whereas the aliphatictri- and multidentate BFCAs produced specific activitiesof 10–20 GBq/µmol. In vitro binding studies of the dif-ferent 99mTc-tricarbonyl labelled octreotide analogueswith AR42 cells revealed significant differences in fa-vour of the neutral complexes (29.9%±2.4% binding for[99mTc(CO)3-Nα-Ac-His-0Tyr3octreotate]0 vs 7.2%±0.6%binding for [99mTc(CO)3-DTPA-0Tyr3octreotate]3–, after4 h at 37°C). On the other hand, the retention of the ac-

In comparison with 111In-DTPA-0Tyr3octreotate,[99mTc(CO)3-His-0Tyr3octreotate]+, [99mTc(CO)3-Nα-Ac-His-0Tyr3octreotate]0 and [99mTc(CO)3-DTPA-0Tyr3oct-reotate]3– provided good images of the 99mTc-labelledcompound except in the case of [99mTc(CO)3-His-0Tyr3octreotate]+ after 4 h p.i. (Fig. 10).

Neurotensin analogues [NT(8–13)] targeting recep-tors expressed on a variety of carcinomas [63] have beenlabelled with 99mTcCO. Eleven NT(8–13) analogueswere functionalised at the N-terminus with an Nα-Achistidine or a histidine similar to 0Tyr3octreotate [13, 64].The almost quantitative formation of uniform and stableproducts was observed at ligand concentrations of 10–5

to 5×10–5 M at 75°C. Unspecific labelling through otherside chains of the peptides was not observed. Althoughthe organometallic core is virtually incorporated in thepeptide, it does not reduce the binding affinity of thepeptide. The KD values of the corresponding labelledpeptides varied between 0.2 nM and 3 nM [native pep-tide NT(8–13): KD=1 nM] [39, 40, 41]. A phase I clinicalstudy has been launched with 99mTcCO-labelled NT de-rivatives.

In the case of other receptor-avid peptides and pro-teins which express an endogenous histidine, such asbombesin or neuropeptide Y, the pronounced avidity ofthe tricarbonyl core for histidine can create problemswith unspecific binding. Pre-labelling procedures can beused to circumvent these problems but this procedure iscumbersome [42]. Garcia-Garayoa et al. could show thatsite-directed post-labelling of bombesin with 99mTcCO ispossible by introduction of an Nα-Ac-histidine (triden-tate chelator) at the N-terminus of the peptide [43]. As aresult, a single, stable species was formed and unspecificlabelling was negligible.

Examples of large proteins labelled with Tc/Re-tricarbonyl

Single-chain antibody fragments (scFvs) have the poten-tial for tumour targeting, since they yield high tumour-to-background ratios at early time points. As scFvs be-come available from combinatorial libraries, a general,efficient and straightforward radiolabelling methodwould be desirable to exploit this technology. Unfortu-nately, there has been no convenient 99mTc labellingtechnique for scFvs. Conventional labelling strategies re-ly on the presence of free sulphuryl groups (via cys-teine), which have to be introduced chemically or geneti-cally. The free cysteines present a problem for routineproduction and storage owing to interference with disul-phide bridges of the scFvs, which favours misfolding ofthe protein. Other procedures use reduced disulphidebridges for “direct labelling” of the protein. As a resultthe biological activity is often lost.

The group at the Paul Scherrer Institute and Plückthun et al. have successfully developed a standard

1537

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Tab

le2.

Com

para

tive

bio

dist

ribu

tion

(%

ID

/g)o

f va

riou

s 0 T

yr3 o

ctre

otat

e de

riva

tive

s fu

ncti

onal

ised

for

labe

llin

g w

ith

99m

Tc-t

rica

rbon

yl a

nd 1

11In

-DT

PA-0

Tyr3

octr

eota

te in

mal

e L

ewis

rats

bea

ring

CA

2094

8 pa

ncre

atic

tum

ours

. For

str

uctu

re o

f B

FC

As

see

Fig

.9

[BF

CA

]-0 T

yr3 o

ctre

otat

e[N

α-A

c-H

is]

[His

][D

TPA

][i

soD

TPA

][I

DA

][1

11In

-DT

PA]

Tim

e p.

i./ti

ssue

30m

in4

h30

min

4h

30m

in4

h30

min

4h

30m

in4

h60

min

4h

Blo

od0.

2±0.

00.

03±

0.0

3.2±

0.1

0.3±

0.0

0.4±

0.0

0.2±

0.0

0.4±

0.1

0.1±

0.0

0.2±

0.0

0.03

±0.

00.

1±0.

00.

01±

0.0

Liv

er0.

5±0.

10.

2±0.

117

.4±

2.4

2.8±

0.2

0.6±

0.3

0.5±

0.1

0.3±

0.1

0.3±

0.1

0.4±

0.1

0.2±

0.0

n.a.

n.a.

Kid

ney

1.9±

0.2

0.7±

0.1

6.1±

0.7

3.2±

1.2

5.3±

1.0

5.7±

2.3

2.2±

1.1

3.2±

1.2

2.2±

0.7

1.1±

0.2

2.7±

0.9

2.3±

0.3

Pan

crea

s8.

5±1.

15.

9±0.

92.

4±0.

32.

5±0.

91.

7±0.

62.

0±0.

51.

8±0.

92.

0±0.

75.

7±2.

05.

8±1.

86.

8±1.

25.

7±1.

4T

umou

r3.

2±0.

41±

0.2

3.5±

0.3

2.2±

0.4

1.1±

0.1

1.2±

0.2

1.1±

0.1

1.2±

0.4

2.7±

0.3

1.1±

0.2

2.7±

0.4

2.3±

0.7

Tum

our/

bloo

d16

331

72.

55

310

1536

2723

0T

umou

r/li

ver

6.5

50.

20.

72

29

47.

56.

5–

–T

umou

r/ki

dney

1.6

1.5

0.6

0.7

0.2

0.2

0.5

0.4

1.2

11

1

labelling protocol employing the 99mTcCO precursor forscFvs and “mini-antibodies” (bi- and trivalent con-structs of scFvs) carrying an N- or a C-terminal His-tag(Fig. 11) [45, 46, 65]. The method is particularly ele-

gant because the His-tag is genetically expressed forease of purification of the protein on a nickel affinitycolumn. Mixing of the scFvs together with 99mTcCO inbuffer at 37°C for 15 min results in >90% incorporationof the total activity (Fig. 12). No protein aggregationcould be detected as determined by size-exclusion chro-matography. With this gentle procedure specific activi-ties of up to 3.3 GBq/mg protein could be achieved.Displacement experiments in the presence of 100-foldexcess of free histidine resulted in only minor dissocia-tion of activity from the labelled scFvs, which provedthe site specificity and stability of the His-tag labelling.ScFvs bearing no His-tag showed only minor incorpo-ration of the radioactivity (16% incorporation of initialactivity). If the protein had endogenous histidines, lessthan 25% of initial activity was incorporated. These da-ta show that labelling occurs predominantly through theHis-tag. Further experiments showed that three histi-dines in a row are sufficient for stable incorporation of99mTcCO. The in vitro immunoreactivity of a series ofseven different labelled scFvs ranged from 57% to 97%(Table 3). The in vivo stability of labelled scFvs wastested with an anti erbB2/Her2 scFv 4D5 in tumour-bearing nu/nu mice. Analysis of the whole-mouse seraafter 1 h revealed that 75% of the injected activity wasstill migrating with the intact scFvs, whereas the rest ofthe activity was bound to albumin and high-molecular-weight proteins. The tumour localisation was 1.4%

1538

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

Fig. 11. Section of a computer-generated model of a proteinlabelled with technetium-tricar-bonyl via His4 and His2 of a 5-His-tag

Fig. 12. Incorporation of 99mTcCO into scFvs with a 5-His-tag(◆◆), with endogenous histidine (●) and with no histidine (■) inbuffer at 37°C. After 60 min the labelled protein was challenged witha 100-fold excess of free histidine. Only 1.5% of initial radioactivitywas released in case of the 5-His-tagged scFvs, compared with 29%in the case of scFvs without histidine in the protein sequence

Table 3. Biological activities ofvarious scFvs after 99mTcCO la-belling

ScFv Antigen Method Activity

MOC31 EGP2 Cell binding 90%4D5 c-erbB2 Cell binding 87%M603-H11 Phosphorylcholine Affinity column 97%FITC-E2 FITC-albumin Gel shift 87%M12 Mucin 44-mer peptide, Dynabeads 57%

i.d./g 24 h post injection, giving rise to tumour/bloodratios of approximately 9. Elevated renal uptake andprolonged retention were detected (108.6%±21.7%i.d./g), which presumably represents an inherent clear-ance property of scFvs. The reason why radioiodinatedscFvs do not show such high renal retention of radioac-tivity is the efficient enzymatic dehalogenation in vivo.Waibel et al. have recently shown that lowering the iso-electric point of the protein, by means of succinylationof the lysine side-chain and co-administration of excessL-lysine, can significantly reduce the high renal uptakeof technetium-tricarbonyl labelled scFvs by a factor ofapproximately 2 [66].

Murray et al. have performed an interesting compara-tive study of reduction-mediated and 188ReCO direct la-belling of anti-MUC1 antibodies in vitro [48]. MUC1mucin is up-regulated and abnormally glycosylated inbladder cancer and is a promising target for intravesicalradioimmunotherapy. The preliminary results clearly re-vealed better retention of immunoreactivity of the188ReCO-labelled mAb (80% at 48 h post labelling)compared with the 2-mercaptoethanol-reduced andRe(V)-labelled mAb (<20% at 48 h post labelling). Thissuggests that the carbonyl labelling methodology may bemore appropriate for intravesical radioimmunotherapyusing 188Re.

Certainly more efforts are required to improve the bi-ological characteristics of Tc/ReCO-labelled antibodiesand scFvs in order to permit routine clinical and eventu-ally therapeutic application. However, the stability andefficiency of this gentle labelling technique for scFvsand antibodies is a decisive advantage.

Miscellaneous examples of organometallic radiolabelled biomolecules

Apart from organometallic technetium and rheniumcomplexes, there have been only a few reports on otherorganometallic compounds of radionuclides for potentialradiopharmaceutical use. Jaouen et al. described the in-sertion of ruthenocene at the 17α-postion of oestradiol[25]. The compound showed moderate retention of bind-ing affinity towards the oestrogen receptor (2% com-pared with native oestrogen). For potential therapeuticapplication, it was suggested to use the β-emitting iso-tope 105Ru (β–, 1.917 MeV, t1/2=4.44 h). The use of orga-nometallic [67Ga]dimethylgallium(III) acetylacetonate(67Ga, electron capture, 1.00 MeV, t1/2=3.26 days) formyocardial imaging agent had to be abandoned becauseof the high accumulation of the compound in the liverand the slow clearance from the blood pool. Reversibleassociation of the (CH3)2[67Ga]+ core with red bloodcells was found to be the reason for the disappointing re-sults [67].

Wenzel et al. have described the potential of 97Ru- or103Ru-ruthenocene-glycine derivatives (“Ru-ruppuran”)

as a surrogate for 123I-hippuran [68] (97Ru: γ, 216 keV,t1/2=2.9 days; 103Ru: γ, 763 keV, t1/2=39.4 days). The ab-sence of β-emission, the suitable γ-energy and the longerhalf-life of the ruthenium isotopes compared with io-dine-123 make them attractive. The renal clearance rateof the organometallic derivatives has been comparedwith that of 125I-hippuran in rabbits. A similar renal andplasma clearance pattern was found for 103Ru-ruppuranand 125I-hippuran. Within the first 20 min p.i., 103Ru-ruppuran was eliminated more rapidly than 123I-hipp-uran, while after 20 min p.i. 125I-hippuran cleared better.This tendency was similar for the plasma clearance. Theauthors claim that the absorbed dose in the bladder andthe kidneys would be slightly lower for 97Ru-ruppuranthan for conventional 123I-hippuran.

Conclusion

It is apparent that organometallic compounds are a valu-able and serious alternative to state of the art labellingtechniques. The encouraging results of preclinical andclinical studies with organometallic labelled, tumouraffine peptides and scFv fragments form the basis forfurther investigations. The potential for use of organo-metallic labelling techniques in nuclear medicine willalso depend on the therapeutic success of organometalliccompounds and the availability of appropriate radionucl-ides. In the future, chemists and radiopharmacists will beequally challenged to exploit the aqueous organometallicchemistry of potential radionuclides to develop noveltechniques and compounds for diagnostic and therapeu-tic application.

Acknowledgements. We thank Mary Dyszlewski, Robert Waibel,Ilse Novak-Hofer for their help in preparing the manuscript.

References

1. Holman BL, Jones AG, Listerjames J, Davison A, Abrams MJ,Kirshenbaum JM, Tumeh SS, English RJ. A New Tc-99m-labelled myocardial imaging agent, hexakis(tert-butylisoni-trile)-technetium(I) Tc-99m Tbi – initial experience in the hu-man. J Nucl Med 1984; 25:1350–1355.

2. Piwnica-Worms D, Rao VV, Kronauge JF, Croop JM. Charac-terization of multidrug-resistance P-glycoprotein transportfunction with an organotechnetium cation. Biochemistry 1995;34:12210–12220.

3. Herman LW, Sharma V, Kronauge JF, Barbarics E, HermanLA, Piwnica-Worms D. Novel hexakis(areneisonitrile)techne-tium(I) complexes as radioligands targeted to the multidrug-resistance P-glycoprotein. J Med Chem 1995; 38:2955–2963.

4. Alberto R, Schibli R, Egli A, Schubiger PA, Herrmann WA,Artus G, Abram U, Kaden TA. Metal-carbonyl syntheses.22.Low-pressure carbonylation of MOCl4

– and MO4– – the tech-

netium(I) and rhenium(I) complexes [NEt4]2[MCl3(CO)3]. J Organomet Chem 1995; 493:119–127.

1539

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

5. Alberto R, Schibli R, Egli A, Schubiger AP, Abram U,Kaden TA. A novel organometallic aqua complex of techne-tium for the labeling of biomolecules: synthesis of[99mTc(H2O)3(CO)3]+ from [99mTcO4]– in aqueous solution andits reaction with a bifunctional ligand. J Am Chem Soc 1998;120:7987–7988.

6. Alberto R, Ortner K, Wheatley N, Schibli R, Schubiger AP. Syn-thesis and properties of boranocarbonate: a convenient in situCO source for the aqueous preparation of [99mTc(H2O)3(CO)3]+.J Am Chem Soc 2001; 123:3135–3136.

7. Schibli R, Schwarzbach R, Ortner K, Schmalle H, Dumas C,Alberto R, Egli A, Schubiger PA. Steps toward high specificactivity labeling of biomolecules for therapeutic application:preparation of precursor [188Re(H2O)3(CO)3]+ and synthesis oftailor-made bifunctional ligand systems. Bioconjugate Chem2002; in press.

8. Top S, Vessieres A, Jaouen G. Synthetic strategy for organo-metallic complexes of rhenium with exceptionally high-affini-ty for the estradiol-receptor – their potential use as imagingand therapeutic agents. J Chem Soc-Chem Commun 1994:453–454.

9. Wenzel M. Tc-99m Labeling of cymantrene-analogs with dif-ferent substituents – a new approach to Tc-99m radiodiagnos-tics. J Label Compd Radiopharm 1992; 31:641–650.

10. Minutolo F, Katzenellenbogen JA. A convenient three-compo-nent synthesis of substituted cyclopentadienyl tricarbonyl rhe-nium complexes. J Am Chem Soc 1998; 120:4514–4515.

11. Minutolo F, Katzenellenbogen JA. Boronic acids in the three-component synthesis of carbon-substituted cyclopentadienyltricarbonyl rhenium complexes. J Am Chem Soc 1998; 120:13264–13265.

12. Winter M. Organorhenium compounds. Gmelin handbook ofinorganic chemistry, vol. 1, 8th edn. Berlin, Heidelberg, NewYork: Springer, 1989.

13. Egli A, Alberto R, Tannahill L, Schibli R, Abram U, Schaffland A, Waibel R, Tourwe D, Jeannin L, Iterbeke K,Schubiger PA. Organometallic Tc-99m-aquaion labels peptideto an unprecedented high specific activity. J Nucl Med 1999;40:1913–1917.

14. Schibli R, La Bella R, Alberto R, Garcia-Garayoa E, Ortner K,Abram U, Schubiger PA. Influence of the denticity of ligandsystems on the in vitro and in vivo behavior of Tc-99m(I)-tri-carbonyl complexes: a hint for the future functionalization ofbiomolecules. Bioconjugate Chem 2000; 11:345–351.

15. Schibli R, Katti KV, Higginbotham C, Volkert WA, Alberto R.In vitro and in vivo evaluation of bidentate, water-solublephosphine ligands as anchor groups for the organometallic fac-99mTc(CO)3

+ core. Nucl Med Biol 1999; 26:711–716.16. Pietzsch HJ, Gupta A, Reisgys M, Drews A, Seifert S,

Syhre R, Spies H, Alberto R, Abram U, Schubiger PA, Johannsen B. Chemical and biological characterization oftechnetium(I) and rhenium(I) tricarbonyl complexes with di-thioether ligands serving as linkers for coupling the Tc(CO)3and Re(CO)3 moieties to biologically active molecules. Bio-conjugate Chem 2000; 11:414–424.

17. Schibli R, Alberto R, Petrig J, Ortner K, Schmalle H, Sigrist B, Stahel J, Schwarzbach R, Schubiger AP. Making thestep from diagnosis to therapy: improved 99mTc/188Re-tricar-bonyl preparation for high specific activity labeling of biomol-ecules. J Label Compd Radiopharm 2001; 44 Suppl 1:S723-S725.

18. Dumas C, Petrig J, Schibli R, Stahel J, Schubiger AP, Spadola L, Scapozza L, Mertens J. Functionalization of glu-

cose for the labeling with 99mTc-tricarbonyl. J Label CompdRadiopharm 2001; 44 Suppl 1:S57–S59.

19. Petrig J, Schibli R, Dumas C, Alberto R, Schubiger PA. Derivatization of glucose and 2-deoxyglucose for transitionmetal complexation: substitution reactions with organo-metallic Tc-99m and Re precursors and fundamental NMRinvestigations. Chemistry – a European Journal 2001; 7:1868–1873.

20. Jaouen G, Top S, Vessieres A, Pigeon P, Leclercq G, Laios I.First anti-oestrogen in the cyclopentadienyl rhenium tricarbon-yl series. Synthesis and study of antiproliferative effects.Chem Commun 2001:383–384.

21. Spradau TW, Katzenellenbogen JA. Ligands for the estrogenreceptor, containing cyclopentadienyltricarbonylrhenium units.Bioorg Med Chem Lett 1998; 8:3235–3240.

22. Wenzel M, Klinge C. Tc-99m-labeled estradiol derivatives –synthesis, organ distribution and tumor affinity. J LabelCompd Radiopharm 1994; 34:981–987.

23. Top S, Elhafa H, Vessieres A, Quivy J, Vaissermann J,Hughes DW, McGlinchey MJ, Mornon JP, Thoreau E,Jaouen G. Rhenium carbonyl-complexes of beta-estradiol de-rivatives with high-affinity for the estradiol-receptor – an ap-proach to selective organometallic radiopharmaceuticals. J AmChem Soc 1995; 117:8372–8380.

24. Le Bideau F, Pérez-Luna A, Marrot J, Rager M-N, Stéphan E,Top S, Jaouen G. New and efficient synthesis of CpRe(CO)3substituted steroids. Tetrahedron 2001; 57:3939–3944.

25. Tang J, Top S, Vessieres A, Sellier N, Vaissermann J,Jaouen G. Synthesis of 17α-ruthenocenyl-17 beta-oestradioland its potential as a radiopharmaceutical agent. Appl Organo-met Chem 1997; 11:771–781.

26. Wust F, Carlson KE, Katzenellenbogen JA, Spies H, Johannsen B. Synthesis and binding affinities of new 17 α-substituted estradiol-rhenium “n+1” mixed-ligand and thio-ethercarbonyl complexes. Steroids 1998; 63:665–671.

27. Wust F, Skaddan MB, Leibnitz P, Spies H, KatzenellenbogenJA, Johannsen B. Synthesis of novel progestin-rhenium conju-gates as potential ligands for the progesterone receptor. BioorgMed Chem 1999; 7:1827–1835.

28. Wenzel M, Klinge C, Saidi M. Tc-99m and deuterium-labeledradiodiagnostics – comparison of HMPAO with cytectrene de-rivatives of cyclic amines. J Label Compd Radiopharm 1993;33:1039–1051.

29. Kuntschke D, Wenzel M, Schulze P. New Tc-99m-cytectreneamine compounds as specific brain imaging agents. J LabelCompd Radiopharm 1995; 36:193–203.

30. Wenzel M, Saidi M. Esters of Tc-99m labeled cyte-ctrenecarboxyxlic acid with alcohols of cyclic amines as cere-bral radiodiagnostic agents. J Label Compd Radiopharm 1993;33:77–80.

31. Cesati RR, Tamagnan G, Baldwin RM, Zoghbi SS, Innis RB,Kula NS, Baldessarini RJ, Katzenellenbogen JA. Synthesis ofcyclopentadienyltricarbonyl rhenium phenyltropanes by dou-ble ligand transfer: organometallic ligands for the dopaminetransporter. Bioconjugate Chem 2002; 13:29–39.

32. Hoepping A, Reisgys M, Brust P, Seifert S, Spies H, AlbertoR, Johannsen B. TROTEC-1: a new high-affinity ligand for la-beling of the dopamine transporter. J Med Chem 1998;41:4429–4432.

33. Tamagnan G, Baldwin RM, Kula NS, Baldessarini RJ, InnisRB. Cyclopentadienyltricarbonylrheniumbenzazepines: syn-thesis and binding affinity. Bioorg Med Chem Lett 2000;10:1113–1115.

1540

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

34. Dyszlewski M, Blake H, Dahlheimer J, Piwnica-Worms D.Characterization of Tc-99m-tricarbonyl complexes as trans-porter substrates of the multidrug resistance (MDR1) P-glyco-protein. J Label Compd Radiopharm 2001; 44 Suppl 1:S63–S65.

35. Maresca KP, Kronauge JF, Babich JW, Carmel A, Marshall M,Jones A. Tc-99m-isonitriles for renal imaging. J Label CompdRadiopharm 2001; 44 Suppl 1:S72–S74.

36. Netter M, Zolle I, Schibli R, Hammerschmidt F, Berger M,Schubiger AP. Synthesis and characterization of [99mTc]meto-midate as the tricarbonyl-conjugate. J Label Compd Radio-pharm 2001; 44 Suppl. 1:S624–S626.

37. Du J, Hiltunen J, Marquez M, Nilsson S, Holmberg AR. Tech-netium-99m labelling of glycosylated somatostatin-14. ApplRadiat Isot 2001; 55:181–187.

38. Marmion M, Alberto R, Bugaj J, Chinen L, Schmidt M, Srinivasan A. Preparation and biodistribution of[99mTc(CO)3His,Tyr]octreotate. J Label Compd Radiopharm1999; 42 Suppl 1:S231–S233.

39. Brühlmeier M, Garcia-Garayoa E, Blanc A, Holzer B, Gergely S, Tourwé D, Schubiger AP, Bläuenstein P. Stabiliza-tion of neurotensin analogues: effect on peptide catabolism,biodistribution and tumor binding. Nucl Med Biol 2002;29:321–327.

40. Garcia-Garayoa E, Allemann-Tannahill L, Bläuenstein P, Willmann M, Carrel-Rémy N, Tourwé D, Iterbeke K, Con-rath P, Schubiger AP. In vitro and in vivo evaluation of newradiolabeled neurotensin(8–13) analogues with high affinityfor NT1 receptors. Nucl Med Biol 2001; 28:75–84.

41. Garcia-Garayoa E, Bläuenstein P, Brühlmeier M, Blanc A,Iterbeke K, Conrath P, Tourwé D, Schubiger AP. Preclinicalevaluation of a new, stabilized neurotensin(8–13) pseudopep-tide radiolabeled with Tc-99m. J Nucl Med 2002; 43:374–383.

42. Langer M, La Bella R, Garcia-Garayoa E, Beck-Sickin-ger AG. 99mTc-labeled neuropeptide Y analogues as potentialtumor imaging agents. Bioconjugate Chem 2001; 12:1028–1034.

43. La Bella R, Garcia-Garayoa E, Bähler M, Bläuenstein P,Schibli R, Conrath P, Tourwé D, Schubiger AP. A 99mTc(I)post-labeled high affinity bombesin analogue as a potentialtumor imaging agent. Bioconjugate Chem 2002; 13:599–604.

44. Baidoo KE, Knight LC, Romano JE, Gabriel JL, Maurer AH.Synthesis and evaluation of new bifunctional chelator de-signed for labeling biomolecules by the technetium tricarbonyltechnology. J Label Compd Radiopharm 2001; 44 Suppl1:S66–S68.

45. Waibel R, Alberto R, Willuda J, Finnern R, Schibli R, Stichelberger A, Egli A, Abram U, Mach JP, Plückthun A,Schubiger PA. Stable one-step technetium-99m labeling ofHis-tagged recombinant proteins with a novel Tc(I)-carbonylcomplex. Nat Biotechnol 1999; 17:897–901.

46. Willuda J, Honegger A, Waibel R, Schubiger AP, Stahel R,Zangemeister-Wittke U, Plückthun A. High thermal stability isessential for tumor targeting of antibody fragments: engineer-ing of a humanized anti-epithelial glycoprotein-2 (epithelialcell adhesion molecule) single-chain Fv fragment. Cancer Res1999; 59:5758–5767.

47. Amann A, Decristoforo C, Ott I, Wenger M, Bader D, Alberto R, Putz G. Surfactant protein B labelled with[99mTc(H2O)3(CO)3]+ retains biological activity in vitro. NuclMed Biol 2001; 28:243–250.

48. Murray A, Scholfield DP, Schibli R, Novak-Hofer I, Waibel R,Vincent RM, Perkins AC. A preliminary comparison of meth-ods for production of 188Re-C595 antibodies for radioimmuno-therapy of bladder cancer. J Label Compd Radiopharm 2001;44: S573–S575.

49. Pietzsch HJ, Johannsen B. Development of technetium-99m-based CNS receptor ligands: have there been any advances?Eur J Nucl Med 2002; 29:263–275.

50. Kung MP, Stevenson DA, Plossl K, Meegalla SK, Beckwith A, Essman WD, Mu M, Lucki I, Kung HF. Tc-99mTRODAT-1: a novel technetium-99m complex as a dopaminetransporter imaging agent. Eur J Nucl Med 1997; 24:372–380.

51. Kung HF, Kim HJ, Kung MP, Meegalla SK, Plossl K, Lee HK.Imaging of dopamine transporters in humans with technetium-99m TRODAT-1. Eur J Nucl Med 1996; 23:1527–1530.

52. Meegalla S, Plössel K, Kung M-P, Stevenson DA, Liable-Sands L, Rheingold AL, Kung HF. First example of a 99m-Tccomplex as a dopamine transporter imaging agent. J Am ChemSoc 1995; 117:11037–11038.

53. Alberto R, Schibli R, Schubiger AP, Abram U, Pietzsch HJ,Johannsen B. First application of fac-[99mTc(H2O)3(CO)3]+ inbioorganometallic chemistry: design, structure, and in vitro af-finity of a 5-HT1A receptor ligand labeled with Tc-99m. J AmChem Soc 1999; 121:6076–6077.

54. Wald J, Alberto R, Ortner K, Candreia L. Aqueous one-potsynthesis of derivatized cyclopentadienyl-tricarbonyl com-plexes of Tc-99m with an in situ CO source: application to aserotonergic receptor ligand. Angew Chem-Int Edit 2001;40:3062–3066.

55. Salmain M, Vessieres A, Brossier P, Butler IS, Jaouen G. Car-bonylmetalloimmunoassay (Cmia) a new type of nonradioiso-topic immunoassay – principles and application to phenobarbi-tal assay. J Immunol Methods 1992; 148:65–75.

56. Jaouen G, Vessieres A, Butler IS. Bioorganometallic chemistry– a future direction for transition-metal organometallic chem-istry. Accounts Chem Res 1993; 26:361–369.

57. Jaouen G, Vessieres A. Transition-metal carbonyl estrogen-re-ceptor assay. Pure Appl Chem 1985; 57:1865–1874.

58. Jaouen G, Vessieres A, Top S, Ismail AA, Butler IS. Metal-carbonyl fragments as a new class of markers in molecular bi-ology. J Am Chem Soc 1985; 107:4778–4780.

59. Skaddan MB, Wust FR, Katzenellenbogen JA. Synthesis andbinding affinities of novel re-containing 7 alpha-substitutedestradiol complexes: models for breast cancer imaging agents.J Org Chem 1999; 64:8108–8121.

60. Decristoforo C, Melendez-Alafort L, Sosabowski JK,Mather SJ. Tc-99m-HYNIC-Tyr3-octreotide for imaging so-matostatin-receptor-positive tumors: preclinical evaluation andcomparison with In-111-octreotide. J Nucl Med 2000;41:1114–1119.

61. Decristoforo C, Mather SJ, Cholewinski W, Donnemiller E,Riccabona G, Moncayo R. Tc-99m-EDDA/HYNIC-TOC: anew Tc-99-labelled radiopharmaceutical for imaging somato-statin receptor-positive tumours: first clinical results and intra-patient comparison with In-111-labelled octreotide derivatives.Eur J Nucl Med 2000; 27:1318–1325.

62. Vallabhajosula S, Moyer BR, ListerJames J, McBride BJ,Lipszyc H, Lee H, Bastidas D, Dean RT. Preclinical evaluationof technetium-99m-labeled somatostatin receptor-binding pep-tides. J Nucl Med 1996; 37:1016–1022.

63. Vincent JP. Neurotensin receptors: binding properties, trans-duction pathways, and structure. Cell Mol Neurobiol 1995;15:501–512.

1541

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002

64. Waibel R, Novak-Hofer I, Schibli R, Blauenstein P, Garcia-Garayoa E, Schwarzbach R, Zimmermann K, PellikkaR, Gasser O, Blanc A, Bruhlmeier M, Schubiger PA. Radio-pharmaceuticals for targeted tumor diagnosis and therapy.Chimia 2000; 54:683–688.

65. Willuda J, Kubetzko S, Waibel R, Schubiger PA, Zangemeister-Wittke U, Plückthun A. Tumor targeting of mono-, di-, andtetravalent anti-p185(HER-2) miniantibodies multimerized byself-associating peptides. J Biol Chem 2001; 276:14385–14392.

66. Stichelberger A, Jager H, Waibel R, Novak-Hofer I, ChesterK, Schubiger AP. Chemical modification of 99mTc-tricarbonyllabeled single-chain antibody fragments for improved in vivobehavior in tumor targeting. J Label Compd Radiopharm2001; 44 Suppl 1:S561–S563.

67. Coggin DK, Mathias CJ, Green MA. Investigation of Ga-67dimethylgallium(III) acetylacetonate as a potential radiophar-maceutical. Nucl Med Biol 1994; 21:283–285.

68. Wenzel M, Meinhold H, Schachschneider G. Ru-labeled ruthenocenoyl-glycine: comparison of the clearance with hippuran. Eur J Nucl Med 1985; 10:138–142.

1542

European Journal of Nuclear Medicine Vol. 29, No. 11, November 2002