99mTc Labeling of Highly Potent Small Peptides - Bioconjugate

Sep 25, 1997 - First Application of fac-[Tc(OH2)3(CO)3] in Bioorganometallic Chemistry: Design, Structure, and in Vitro Affinity of a 5-HT1A Receptor ...
3 downloads 8 Views 391KB Size
Bioconjugate Chem. 1997, 8, 621−636

621

REVIEWS 99mTc

Labeling of Highly Potent Small Peptides

Shuang Liu,* D. Scott Edwards, and John A. Barrett Radiopharmaceuticals Division, The DuPont Merck Pharmaceutical Company, 331 Treble Cove Road, North Billerica, Massachusetts 01862. Received April 21, 1997 INTRODUCTION

Peptides are compounds that contain amino acids (Raminocarboxylic acids) linked by amide (peptide) bonds. Designed by nature for stimulating, inhibiting, or regulating numerous life functions, peptides for a long time have been considered ideal agents for therapeutic applications (1). Although the 111In-labeled peptide was first explored for use in nuclear medicine in 1981 (2), it was not until recently that 99mTc-labeled peptides have emerged as an important class of radiopharmaceuticals in diagnostic nuclear medicine. Of particular interest are 99mTc-labeled chemotactic peptides (3-11), small peptides based on platelet factor 4 (12), and tuftsin receptor antagonists (13-15) for imaging focal sites of infection, somatostatin analogs for imaging tumors (16-20), and platelet glycoprotein IIb/IIIa (GPIIb/IIIa) receptor antagonists for imaging thrombi (21-34). In the past two decades, monoclonal antibodies (MAbs) or their fragments, F(ab′) or F(ab′)2, have been widely used as vehicles to carry radionuclides of diagnostic or therapeutic characteristics to a specific target such as a tumor (35-40). Although considerable progress has been made in this area (40-43), clinical studies with radiolabeled MAbs have often demonstrated limited accumulation in the tumor, and relatively slow blood clearance due to their high molecular weight, resulting in only modest target-to-background ratios. The poor match between the pharmacokinetics and the physical half-life of 99mTc has limited the development of 99mTc-labeled MAbs and their fragments as commercial target-specific radiopharmaceuticals. The difference between proteins and peptides is their sizes. The term “peptides” is usually used to refer to those compounds containing fewer than 100 amino acids with a molecular mass of about 10 000 Da (44). Small peptides refers to peptides with fewer than 30 amino acids or molecular mass of 70% of pulmonary emboli arise from DVT in the lower extremities (132). However, existing diagnostic modalities are inadequate to diagnose and determine the morphology of the evolving thrombus (133-137). Thus, the development of agents that will not only detect the location but, in addition, determine the age of the thrombi is a critical unmet need in diagnostic nuclear medicine. A venous thrombus is an intravascular deposit predominantly comprising fibrin, and aggregates of platelets and red blood cells. Platelet aggregation is mediated by fibrinogen, which binds via the Arg-Gly-Asp (RGD) tripeptide sequence to the GPIIb/IIIa receptor expressed on activated platelets. Much of the early work in thrombus imaging was focused on the radiolabeling of fibrinogen (138, 139). In 1976, it was reported that platelets can be labeled very efficiently with [111In]oxine (140). Since then, 111In-labeled platelets have been used to detect left ventricular thrombi (141, 142), pulmonary emboli (143), and DVT (144, 145). However, imaging using 111In-labeled platelets suffers from several limitations. The technique is too complex and time-consuming for routine clinical use (146). It usually takes 24-72 h to get useful images. The 111In-labeled platelets cannot detect small thrombi such as coronary artery thrombi due to the relatively high background (blood pool) activity (147). The slow blood clearance and poor T/B severely limit the use of 111In-labeled platelets. Platelet activation and deposition are the initial events in acute thrombus formation and may continue for a variable period as thrombus organization proceeds (31). Activated platelets express the GPIIb/IIIa receptor, which recognizes proteins and peptides bearing the RGD tripeptide sequence, while nonactivated platelets express virtually none of the receptor in its active conformation. A number of RGD-containing small peptides (33, 148151), which are antagonists of the platelet GPIIb/IIIa receptor, have been synthesized and studied for their antithrombotic activities. These small peptides represent a rapidly growing class of antithrombotics. Recently, DeGrado and co-workers (152-155) reported a series of very potent cyclic GPIIb/IIIa receptor antagonists, two (I and II) of which are shown in Figure 3. Peptides I and II have very high selectivity and binding affinity for the GPIIb/IIIa receptor, with IC50 values in the nanomolar range for inhibition of both platelet aggregation and fibrinogen binding (153). This makes them candidates of choice as targeting molecules to carry

Figure 3. Cyclic GPIIb/IIIa receptor antagonists.

the radionuclide (99mTc) to thrombi. Since the RGD tripeptide sequence is vital for maintaining biological activity, the functionalized cyclic peptides III-V were prepared by incorporation of a lysine residue between the arginine residue and the Mamb [mamb ) m-(aminomethyl)benzoic acid] or by attachment of a 6-aminocaproic acid linker on the benzene ring of the Mamb moiety (156, 157). The use of the 6-aminocaproic acid linker keeps the Tc center far from the RGD sequence to minimize the effect of 99mTc labeling on the receptor binding affinity. Since the GPIIb/IIIa receptor is expressed only on activated platelets, the 99mTc-labeled cyclic GPIIb/IIIa receptor antagonists should only be bound to platelets intimately involved in the thrombolic event. The rapid blood clearance of small peptides allows the use of 99mTc and permits earlier imaging of rapidly growing thrombi. The preformed chelate approach has been used to label several cyclic GPIIb/IIIa receptor antagonists with 99mTc (25). It was found that both the cyclic peptide and the BFCA contribute to the differences in physical and biological properties of the [99mTc]BFCA-peptide complex (25, 26). In a canine arteriovenous shunt (AV shunt) model four complexes, [99mTcO(L1-III)]-, [99mTcO(L2III)]-, [99mTcO(L1-V)]-, [99mTcO(L2-V)]-, were found to be incorporated into the growing thrombus under both arterial and venous conditions. The differences in the uptake of these complexes under both arterial and venous conditions are related to the receptor binding affinity of the peptide and the rate of clearance of technetium complexes from the blood circulation. In a canine DVT model, the complex [99mTcO(L1-V)]- was incorporated in the growing thrombus with images clearly detectable within 15 min postinjection. At 2 h postinjection, thrombus-to-blood and thrombus-to-muscle ratios were approximately 7:1 and 10:1, respectively. This clearly demonstrated that the complex [99mTcO(L1-V)]- has the potential for rapid diagnosis of thromboembolic events occurring under both arterial and venous conditions. The indirect labeling approach (Chart 3) has been used to synthesize the complex [99mTcO(L1-V)]- (117). The synthesis of the complex [99mTcO(L1-V)]- was carried out using the ligand exchange method, in which the (EOE)2H2L1-V conjugate was allowed to react with [99mTc]glucoheptonate. It usually requires at least 200 µg of (EOE)2H2L1-V conjugate in each reaction vial to achieve a successful 99mTc labeling. This is consistent with the low labeling efficiency of N2S2 diamidedithiols.

626 Bioconjugate Chem., Vol. 8, No. 5, 1997 Chart 4.

Liu et al.

99m

Tc Labeling of HYNIC-V

Figure 4. Proposed structures for ternary ligand technetium complexes.

HYNIC was also used to label the peptide V with 99mTc (24) (Chart 4). The advantage of using HYNIC as the BFCA is its high labeling efficiency and the choice of various coligands, which allows control of the hydrophilicity and pharmacokinetics of the 99mTc-labeled small peptides. Among various coligands, tricine gives the best radiolabeling efficiency. For example, very high specific activity (e20 000 mCi/µmol) can be achieved for the complex [99mTc(HYNIC-V)(tricine)2]. However, it was found that the complex [99mTc(HYNIC-V)(tricine)2] is not stable, particularly in dilute solutions, and exists as multiple species, which we have attributed to different bonding modalities of the hydrazine moiety of the HYNIC-V and the tricine coligands. Although the animal studies in a canine arteriovenous (AV) shunt and a DVT model show that [99mTc(HYNIC-V)(tricine)2] is able to image both arterial and venous thrombi (23, 29), it would still be difficult to develop for clinical use because of the solution instability and the presence of the many isomeric forms. To prepare [99mTc]HYNIC-V complexes with higher solution stability and less isomerism, EDDA (ethylenediamine-N,N′-diacetic acid), which is more symmetrical and potentially tetradentate, was used as the coligand for the radiolabeling (Chart 4). It was found that EDDA forms a complex, [99mTc(HYNIC-V)(EDDA)], with much higher solution stability and less isomerism (24). However, the HPLC data showed that the complex [99mTc(HYNIC-V)(EDDA)] exists in at least three isomeric forms. In an alternative approach (Figure 4), a water soluble phosphine (TPPTS, TPPDS, and TPPMS) is used as an additional coligand to form a ternary ligand system (27). These monodentate phosphines react readily with [99mTc(HYNIC-V)(tricine)2], replace one of its two tricine coligands, and form complexes, [99mTc(HYNIC-V)(tricine)(L)] (RP444, L ) TPPTS; RP445, L ) TPPDS; RP446, L )

Figure 5. Representative DVT images of complexes RP444, RP445, and RP446 at 15, 60, and 120 min postinfusion. The bar to the right of the images indicates the scale from 0 (white) to 506 (greatest/black). The images have not been filtered.

TPPMS) in high yield and high specific activity (g20 000 Ci/mmol). RP444, RP445, and RP446 are formed as equal mixtures of two isomeric forms, which is due to the resolution of diastereomers resulting from the chiral centers on the peptide backbone and the chirality of the technetium chelate (158). The composition of these complexes was determined to be 1:1:1:1 for Tc:HYNICV:L:tricine through a series of mixed ligand experiments on the tracer (99mTc) level. This composition is maintained over a wide range of relative ligand ratios. In the canine AV shunt model (28), RP444, RP445, and RP446 were adequately incorporated into the arterial and venous portions of the growing thrombus (7.8-9.9 and 0.2-3.7% ID/g, respectively). Figure 5 shows representative unfiltered images of RP444, RP445, and RP446 in the DVT imaging model at 15, 60, and 120 min postinfusion. All three complexes had thrombus uptake (%ID/g ) 2.86 ( 0.4, 3.4 ( 0.9, 3.38 ( 1.1 for RP444, RP445, and RP446, respectively) that far exceeded that of the negative control, [99mTc]albumin. They also show similar blood clearance with a t1/2 values of approximately 90 min. Visualization of DVT can be as early as 15 min postinjection and improves over time with the thrombus/ muscle ratios of 9.7 ( 1.9, 13.8 ( 3.6, and 9.4 ( 2 for RP444, RP445, and RP446, respectively, at 120 min postinjection. The administration of RP444, RP445, and RP446 did not alter platelet function, hemodynamics, or the coagulation cascade. Therefore, all three complexes have the capability to detect rapidly growing venous and arterial thrombi. RP444 was selected for clinical studies

Reviews

as a new thrombus imaging agent for both arterial thrombi and DVT. Obviously, the use of this new ternary ligand system offers several major advantages. First, bonding of the phosphine coligand to the Tc dramatically reduces the number of isomeric forms of the [99mTc]HYNIC-V complexes. Second, the solution stability of [99mTc]HYNICV complexes is dramatically improved. RP444 was found to be stable for at least 6 h under dilute conditions, while the HPLC-purified corresponding 99Tc analog, [99Tc(HYNIC-V)(tricine)(TPPTS)], remains stable in aqueous solution for more than 6 months without any decomposition (158). It is amazing that three different ligands (HYNIC-V, tricine, and phosphine) combine with Tc and form technetium complexes with only two detectable isomers and with extremely high solution stability. Finally, the hydrophilicity of [99mTc]HYNIC-V complexes with this ternary ligand system can be tuned either by altering the number of sulfonato groups or by using water soluble phosphines with other polar functionality. The tricine coligand can also be substituted by other potentially tetradentate aminocarboxylates such as dicine [N-bis(hydroxymethyl)methylglycine] and bicine [N,Nbis(hydroxyethyl)glycine]. However, the specific activity of [99mTc]HYNIC-V complexes using dicine and bicine as coligands is not as high as that of the corresponding tricine complexes. This is consistent with the literature results reported by Abrams and co-workers (93-96), who found that the [99mTc]tricine precursor complex has the highest reactivity with hydrazines. Using the combination of a polydentate aminocarboxylate and a phosphine ligand, HYNIC-conjugated peptides and other HYNICderivatized small molecules can be readily labeled with 99mTc in high specific activity and high stability for potential use as radiopharmaceuticals. The technetium oxidation state and the exact nature of bonding between these three ligands (HYNIC, tricine, and phosphine) and the Tc are not yet clear. It has been proposed that the technetium oxidation state in the [99mTc]tricine complex is 5+ (92, 93). However, the oxidation state might change when the HYNIC group and phosphine coligand are bonded to the Tc center. Furthermore, the technetium oxidation state largely depends on how the HYNIC group binds to the Tc and how one counts the charge on the HYNIC ligand. Complexes containing Tc-hydrazido and Tc-diazenido bonds have been previously reported and characterized by X-ray crystallography (159-165). Recently, Davison and co-workers (166, 167) reported a series of technetium(III) and rhenium(III) complexes of 2-hydrazinopyridine. It was found that unlike phenylhydrazine, 2-hydrazinopyridine has several bonding modalities: neutral bidentate pyridyldiazene (166, 167), neutral monodentate pyridiniumdiazenido (166), and anionic monodentate pyridyldiazenido (167). Various Tc/Re complexes can be prepared depending upon the starting material and reaction conditons. For example, the reaction of NH4[TcO4] with 2-hydrazinopyridine dihydrochloride gives [TcCl3(HNdNC5H4N)(NdNC5H4NH)]. The reaction of [ReCl3(HNdNC5H4N)(NdNC5H4NH)] with triphenylphosphine (PPh3) in the presence of a base such as diisopropylethylamine produces the complex [ReCl2(PPh3)(NdNC5H4N)(NdNC5H4NH)], in which the NdNC5H4N moiety was found to be an anionic monodentate pyridyldiazenido (166). We also found that NH4[TcO4] or [n-Bu4N][TcOCl4] reacts with HYPY‚2HCl in the presence of excess PPh3 to give the complex [Tc(NdNC5H4N)(PPh3)2Cl2]. The NMR data suggest that the NdNC5H4N moiety is likely bidentate pyridyldiazenido (158). When the HYNIC binds to the Tc, it likely forms either

Bioconjugate Chem., Vol. 8, No. 5, 1997 627

a terminal Tc-diazenido or Tc-hydrazido bond. If one assumes that the coordination geometry around the Tc in the ternary ligand complexes is distorted octahedral, the tricine coligand is expected to be tetradentate with the carboxylate O, amine N, and hydroxy O occupying equatorial positions and one of the two remaining hydroxy O’s occupying the apical site of the Tc (Figure 4). Efforts are continuing to better understand the coordination chemistry of this unique, versatile, and complex ternary ligand system. Lister-James and co-workers (31-34) also used 99mTclabeled IIb/IIIa receptor antagonists for the development of new thrombus imaging agents. P280 is a small oligopeptide made of two identical, linked, cyclic 13 amino acid monomers. Each monomer contains a (S-aminopropylcysteine)-Gly-Asp tripeptide sequence, which is mimetic of the RGD sequence found in molecules such as fibrinogen and fibronectin. P280 has high affinity for the GPIIb/IIIa receptor with an IC50 of 87 nM for inhibition of human platelet aggregation (31). Each monomer contains a Cys(Acm)-Gly-Cys(Acm) tripeptide sequence, which forms an N2S2 diamidedithiol bonding unit for 99mTc labeling. Because of the low labeling efficiency of N2S2 diamidedithiols and the presence of protecting group on the two thiol groups, the 99mTc labeling of P280 requires the use of a large amount of peptide (g250 µg) and heating at 100 °C for 15 min when prepared by ligand exchange with [99mTc]glucoheptonate. The specific activity for the [99mTc]P280 complex is low (60 mCi/µmol) compared to that of RP444 (g20 000 mCi/µmol). [99mTc]P280 was reported to give excellent images of DVT in a canine DVT model. However, the thrombus uptake at 4 h postinjection is only 0.0059 ( 0.0025 %ID/g (31), which is much lower than that of RP444 (2.8 ( 0.4 %ID/g) (28). The low thrombus uptake is most likely related to the lower binding affinity of P280 (IC50 ) 87 nM) for the GPIIb/IIIa receptor and faster blood clearance 99mTcP280 (t1/2R ) 1.6 min and t1/2β )20 min) as compared to the binding affinity of peptide V (IC50 ) 6 nM) and the blood clearance of RP444 (t1/2 ) 90 min). P748 is another platelet GPIIb/IIIa receptor antagonist under investigation for imaging pulmonary embolism (33, 44). P748 has a higher receptor binding affinity (IC50 ) 28 nM against aggregation of human platelets) than P280 (IC50 ) 87 nM), and [99mTc]P748 has slower blood clearance with t1/2R ) 9.6 min and t1/2β ) 145 min. It is not surprising that [99mTc]P748 shows a higher uptake (%ID/g ) 0.018 ( 0.0068 at 4 h postinjection) and a higher T/B than [99mTc]P280. Unlike P280, P748 uses an N3S aminediamidethiol chelating unit. Because of the asymmetric character of the chelator, two epimers are seen, one of which is predominant. It was reported that P748 can be easily labeled by ligand exchange with [99mTc]glucoheptonate at room temperature. The reported highest specific activity for [99mTc]P748 is about 2000 mCi/µmol. Apparently, the presence of the amine N donor and a unprotected thiol in the chelator dramatically improves the radiolabeling efficiency. Infection/Inflammation Imaging. Imaging focal sites of infection or inflammation using radiolabeled peptides is another important area in diagnostic nuclear medicine. Accurate and rapid localization of infectious and inflammatory foci facilitate elucidation of the cause of disease and the implementation of a tailored therapeutic regimen (169). Current imaging procedures such as X-ray computed tomography (CT), ultrasound (US), conventional radiography, and magnetic resonance imaging (MRI) rely primarily on focal changes in tissue density to define lesions. In the early stage of an inflammatory lesion, lesion localization using these pro-

628 Bioconjugate Chem., Vol. 8, No. 5, 1997

cedures may be difficult because the tissue changes associated with necrosis have not occurred (6). For a number of years, imaging of inflammation and infection has been performed using either [67Ga]citrate or 111In-labeled white blood cells (WBCs) (170, 171). 99mTc-labeled nonspecific polyclonal IgG’s were also used for detection of infection foci (92, 94, 172-177). Although these agents are well-accepted and efficacious, it usually takes 24 h to obtain diagnostically useful images. Recently, two new radiopharmaceuticals were introduced for imaging sites of infection: [99mTc]hexamethylpropylene amine oxime (HMPAO)-WBCs (178-180) and [99mTc]albumin colloid-WBCs (181). Imaging using both agents can be completed on the same day, but these procedures may impose significant risks to laboratory personnel and patients, particularly with the increasing prevalence of human immunodeficiency virus in the population (6, 182, 183). WBCs, particularly polymorphonuclear leukocytes (PMNLs) and monocytes, accumulate in high concentrations at sites of infection. Therefore, most attention has been directed toward radiolabeling small molecules that bind to both circulating granulocytes and leukocytes. These include chemotactic peptides, analogs of N-formylmethionylleucylphenylalanine (fMLF) (3-11), small peptides based on platelet factor 4 (12), and tuftsin receptor antagonists (13-15). The chemotactic peptide fMLF is a bacterial product that initiates leukocyte chemotaxis by binding to highaffinity receptors on inflammatory cells. Fischman and co-workers (4-11) have investigated a series of radiolabeled chemotactic peptides as infection/inflammation imaging agents. These peptides were modified with HYNIC (for 99mTc labeling) or DTPA (for 111In labeling) at the C terminus. The 99mTc labeling of HYNIC-modified chemotactic peptides can be achieved using various coligands such as glucoheptonate, mannitol, and glucamine (5). It was reported that very high specific activity (g20 000 mCi/µmol) could be achieved using glucoheptonate as the coligand. Different biodistributions were observed for various coligands (5). Animal studies have shown evidence of binding to leukocytes in vivo and localization at sites of infection (6-11). In a rabbit infection model, a 99mTc-labeled fMLF analog gave a target to background ratio greater than or equal to that achieved with [111In]WBCs (11). We also labeled a chemotactic peptide-HYNIC conjugate (fMLFK-HYNIC) using tricine or glucoheptonate as coligand (183). It was found that tricine and glucoheptonate form technetium complexes, [99mTc(fMLFKHYNIC)(L)2] (L ) tricine and glucoheptonate), with many isomeric forms. These technetium complexes are not stable in the absence of excess coligand. The combination of tricine and TPPTS forms a stable ternary ligand complex [99mTc(fMLFK-HYNIC)(tricine)(TPPTS)], which was evaluated in two animal (guinea pig and rabbit) models of focal infection. It was found that the ternary ligand complex was superior with a T/B of 5-7:1 at 4 h postinjection, reflecting an increased clearance from the normal muscle. In the rabbit infection model a transient decrease in WBC count of 35% was observed in all three groups during the first 30 min postinjection (183). A similar neutropenic response was also reported for the 99m Tc-labeled chemotactic peptides, [99mTc]RP050 and 99mTc]RP056 (184). [ Apparently, the 99mTc labeling of these highly potent agonists suffers a drawback, severe reduction of peripheral leukocyte count because of the unlabeled peptide even at very low doses. There are two approaches to avoid this problem. The first approach is to separate the

Liu et al.

radiolabeled peptide from the excess unlabeled peptide using HPLC. This results in a product almost at its theoretical specific activity but is inconvenient for routine clinical use. The alternative approach is to use antagonist peptides, which bind to the receptor but do not trigger the neutropenic effect, as targeting molecules. However, the receptor binding affinities of the antagonists tested to date appear to be much lower than that of the agonists (185). Platelet factor 4 (PF-4) is a 29 kDa homotetrameric protein for which receptors have been identified on PMNLs, monocytes, endothelium, fibroblasts, and hepatocytes. Peptide P483 (acetyl-Lys-Lys-Lys-Lys-Lys-CysGly-Cys-Gly-Gly-Pro-Leu-Tyr-Lys-Lys-Ile-Ile-Lys-LysLeu-Leu-Glu-Ser) is an analog of the C-terminal tridecapeptide of human PF-4. It contains a Cys-GlyCys tripeptide chelating unit for 99mTc labeling and a pentalysine sequence on the N terminus to promote renal clearance (12). It was reported that complexation of [99mTc]P483 with heparin substantially enhanced the binding to WBCs and resulted in improved uptake in sites of infection in a rabbit infection model (12). The in vitro distribution in human blood suggests that [99mTc]P483H associates with specific WBCs, particularly monocytes. In the rabbit infection model, [99mTc]P483H showed slightly higher infection uptake (0.062 ( 0.022 %ID/g) than [111In]oxine-WBCs (0.051 ( 0.008 %ID/g), and a 6-fold higher T/B, probably due to the rapid blood clearance (12). Tuftsin is a tetrapeptide (TKPR) derived from the Fc portion of IgG. It promotes phagocytosis and chemotaxis of neutrophils and monocyte/macophages. Recently, a 99mTc-labeled tuftsin receptor antagonist [Pic-SC(Acm)GTKPPR; Pic ) picolinic acid] was used for imaging inflammation (13). The Pic-SC(Acm)G sequence forms an N3S chelating unit for 99mTc bonding. Radiolabeling was achieved by ligand exchange with [99mTc]glucoheptonate. In rats with infectious (Escherichia coli) inflammation (13), the 99mTc-labeled tuftsin antagonist was able to give excellent images with the T/B of 3.6, 5.0, and 16.2 at 0.5, 3, and 17 h postinjection, respectively. Another 99mTc-labeled tuftsin receptor antagonist is [99mTc]RP128 [RP128 ) dimethylGSC(Acm)G-TKPPR]. In an inflammatory bowel disease (IBD) model, [99mTc]RP128 showed much better imaging quality than [111In]oxine-WBCs (14). The target (inflamed terminal colon) to background (proximal noninflamed colon) ratios of 2.14, 2.51, 2.90, and 1.90 were obtained at 0.5, 1, 3, and 18 h postinjection, respectively. Both agents were rapidly excreted via the renal system. Tumor Imaging. There is an unmet need for the development of new target-specific tumor-imaging agents. Radiolabeled receptor-based biomolecules such as small peptides are of particular interest because they have the potential to detect primary sites, identify occult metastatic lesions, guide surgical intervention, stage tumors, and predict efficacy of therapeutic agents. When labeled with a suitable radionuclide, they can also be used as radiotherapeutic agents. The peptide that has attracted greatest interest is somatostatin, a tetradecapeptide which exhibits an inhibitory effect on the secretion of numerous hormones, including growth hormone, thyrotrophin, insulin, glucogon, vasoactive intestinal peptide (VIP), and secretin. Somatostatin receptors are overexpressed on a number of human tumors and their metastases (186-189), thereby serving well as the target for tumor imaging. Although the first report of in vivo imaging with a somatostatin analog appeared in 1976 (190), further development was hampered due to rapid degradation of the native peptide

Bioconjugate Chem., Vol. 8, No. 5, 1997 629

Reviews

by plasma and tissue proteases (191). For this reason, analogs of somatostatin have been synthesized using D-amino acids to prolong the in vivo half-life by inhibiting the action of amino and carboxypeptidases. Octreotide (Sandostatin, SMS 201-995) is a metabolically stable analog of somatostatin and has been successfully used for the treatment of acromegaly and GEP tumors (192194). Derivatives of octreotide have been radiolabeled with γ-emitting radionuclides such as 123I (195, 196) and 111In (197-204), and these radiotracers have been successfully used to detect somatostatin receptor-positive tumors by γ-scintigraphy. [111In]DTPA-octreotide (OctreoScan) has been approved in many countries in Europe and North America and has become one of the most commonly used radiopharmaceuticals in clinical tumor imaging. However, this agent suffers several drawbacks such as the long half-life (t1/2 ) 67 h, γ ) 173 keV, 89%, and 247 keV, 94%) and the high cost of 111In. For diagnostic purposes, 99m Tc is more desirable because of its low cost, easy availability, and ideal nuclear characteristics, which better match the rapid blood clerance and fast tumor accumulation of octreotide. In the past several years, various somatostatin analogs have been labeled with 99mTc using different BFCAs, including N2S2 diamidedithiols (19, 20), N3S triamidethiols (19, 20), N3S aminediamidethiols (19, 20), PnAO (propyleneamine oxime) (16), and tetraamines (17). Of particular interest are 99mTc-labeled peptides P587 and P829. Both peptides contain the Tyr-(D-Trp)-Lys-Val sequence, which is responsible for somatostatin receptor binding. The cyclic peptide backbone does not contain the S-S disulfide linkage and is not susceptible to reductive cleavage. In P587, the Gly-Gly-Cys tripeptide constitutes an N3S triamidethiol chelator for 99mTc bonding, while P829 uses the (β-Dap)-Lys-Cys tripeptide sequence to form an N3S aminediamidethiol chelating unit. Studies in CA20948 tumor-bearing rats showed that the tumor uptake of [99mTc]P587 and [99mTc]P829 is comparable to or better than that of [111In]DTPA-octreotide (19). [99mTc]P829 is excreted predominantly through the renal system and has been selected for clinical trials. HYNIC was also used to label somatostatin analogs with 99mTc (205, 206). It was conjugated to the N terminus of octreotide either directly or via a β-alanine or diaminobutanesuccinyl linker. The use of tricine as coligand produced a very high specific activity (∼6000 mCi/µmol). In a tumor bearing mouse model (AR4-2J), all three 99mTc-labeled HYNIC-octreotide conjugates showed comparable tumor uptake to [111In]DTPA-octreotide (205, 206). The observed high blood activity is probably related to the instability of [99mTc]HYNICoctreotide-tricine complexes and plasma protein binding. Somatostatin analogs (e.g. BIM-23014, RC-160, and Sandostatin), which contain a S-S disulfide bond in the cyclic peptide backbone, have also been radiolabeled by the direct labeling approach (1, 18, 207, 208). It is believed that when the S-S bond is reduced, the thiolate S atoms bond to the Tc center and form stable 99mTc complexes (18). It was also reported that these 99mTclabeled somatostatin analogs have not shown any apparent loss of biological activity, nor have they shown any abnormal blood clearance in experimental animals compared to that with [111In]DTPA-octreotide (1, 207, 208). However, several critical questions remain to be answered for this approach. First, how many peptides are bonded to the Tc center? If only one peptide binds to the Tc, which tripeptide sequence (Cys-AA-AA or AAAA-Cys) is involved in Tc bonding? How many 99mTc

species are in the radiolabeled kit? What is the impact of 99mTc labeling on the receptor binding affinity of the cyclic peptide? CONCLUSIONS

From the discussion above, it is apparent that significant progress has been made in the development of peptide-based target specific radiopharmaceuticals. 99mTc labeling of biologically active peptides stemmed from the studies of 99mTc-labeled antibodies for diagnosis or therapy of various tumors. In a very short period of time, 99mTclabeled peptides have become an important class of imaging agents for the detection of not only tumors but also thrombosis and infection/inflammation. 99mTclabeled peptides such as RP444, [99mTc]P280, [99mTc]P483H, [99mTc]P748, [99mTc]P829, and [99mTc]RP128 are now being evaluated in clinical trials. Results from preeclinical studies have lived up to their expectations: high specificity, high uptake in the target organ, and high T/B due to rapid blood clearance of the 99mTc-labeled small peptides. Hopefully, some of these 99mTc-labeled small peptides will soon become commercial products and serve the nuclear medicine community for decades. Radiolabeled small peptides continue to attract interest because of their favorable physical and chemical characteristics compared to antibodies and their fragments. Small peptides, once the receptor binding sequence is identified, can be easily synthesized and modified according to their pharmacokinetic requirements. Ideally, a peptide-based radiopharmaceutical should have the following characteristics: rapid uptake by target tissues, metabolic stability, and fast renal clearance. This is particularly important for infection and tumor imaging because rapid renal clearance minimizes the accumulation of radioactivity in the abdominal area and results in improved T/B. The isotope of choice for radiolabeling will continue to be 99mTc rather than 111In for diagnostic radiopharmaceuticals because of cost and convenience. The experience learned from 99mTc labeling of small peptides can be applied to other highly potent receptor binding small molecules. In addition, these biologically active small peptides can also be used for the development of therapeutic radiopharmaceuticals when labeled with appropriate radionuclides such as 186Re. In the area of tumor imaging, 99mTc-labeled substance P (SP) and vasoactive intestinal peptide (VIP) as well as their analogs may become new classes of tumor-imaging agents. [111In-DTPA-Arg1]SP has been successfully used to visualize SP-positive processes such as adjuvant arthritis and atransplantable pancreatic tumor (209). Studies using [123I]VIP have clearly demonstrated its ultility in localizing intestinal adenocarcinoma and endocrine tumors as well as metastatic tumor sites in humans (210). Of course, the next logical step is 99mTc labeling of substance P (SP) and vasoactive intestinal peptide (VIP) analogs with favorable pharmacokinetics. 99mTc labeling of highly potent small peptides is different from that of simple organic chelators. Because of the possible side-effects, the BFCA-peptide conjugate has to be labeled at very low concentrations (usually 5-20 µg/mL, corresponding to 5 × 10-6 to 2 × 10-5 M for a BFCA-peptide conjugate with molecular mass of 1000 Da and IC50 values in the nanomolar range) to achieve high specific activity. One of the challenges for inorganic chemists is to design new ligand systems that have very high labeling efficiency and form technetium complexes with minimal isomerism. It should be noted that the development of new 99mTclabeling technologies and synthesis of new biologically active peptides with high receptor binding affinities are

630 Bioconjugate Chem., Vol. 8, No. 5, 1997

equally important. Improvement of the pharmacokinetics of 99mTc-labeled peptides can be achieved by modification of both the peptide and the Tc chelate. The development of 99mTc-labeled peptide radiopharmaceuticals is a multidisciplinary effort and requires the collaboration from scientists in several areas, including organic chemistry, inorganic chemistry, biochemistry, biology, formulation chemistry, and nuclear medicine. As Jurrisson (211) stated: “Without their joint efforts nuclear medicine would not be where it is today, nor will it progress”. ACKNOWLEDGMENT

We give special thanks to Dr. Joseph L. Glajch and Dr. Steven B. Haber for helpful discussions. LITERATURE CITED (1) Thakur, M. L. (1995) Radiolabeled peptides: now and future. Nucl. Med. Commun. 6, 724-732. (2) Zoghbi, S. S., Thakur, M. L., and Gottschalk, A. (1981) Selective cell labeling: a potential radioactive agent for labeling of human neutrophils. J. Nucl. Med. 22, P32 (abstract). (3) Baidoo, K. E., Stathis, M., Scheffiel, U., Lever, S. Z., and Wagner, H. N., Jr. (1993) High affinity Tc-labeled chemotactic peptides. J. Nucl. Med. 34, 18P (abstract 62). (4) Babich, J. W., Solomon, H., Pike, M. C., Kroon, D., Graham, W., Abrams, M. J., Tompkins, R. G., Rubin, R. H., and Fischman, A. J. (1993) Technetium-99m- labeled hydrazino nicotinamide derivatized chemotactic peptide analogs for imaging focal sites of bacterial infection. J. Nucl. Med. 34, 1967-1974. (5) Babich, J. W., and Fischman, A. J. (1995) Effect of “coligand” on the biodistribution of 99mTc-labeled hydrazino nicotinic acid derivatized chemotactic peptides. Nucl. Med. Biol. 22, 25-30. (6) Fischman, A. J., Babich, J. W., and Rubin, R. H. (1994) Infection imaging with technetium-99m-labeled chemotactic peptide analogs. Semin. Nucl. Med. 24, 154-168, and references cited therein. (7) Fischman, A. J., Babich, J. W., and Strauss, H. W. (1993) A ticket to ride: peptide radiopharmaceuticals. J. Nucl. Med. 34, 2253-2263. (8) Fischman, A. J., Rauh, D., Solomon, H., Babich, J. W., Tompkins, R. G., Kroon, D., Strauss, H. W., and Rubin, H. R. (1993) In vivo bioactivity and biodistribution of chemotactic peptide analogs in nonhuman primates. J. Nucl. Med. 34, 2130-2134. (9) Babich, J. W., Graham, W., Barrow, S. A., and Fischman, A. J. (1995) Comparison of the infection imaging properties of a 99mTc labeled chemotactic peptide with 111In IgG. Nucl. Med. Biol. 22, 643-648. (10) Babich, J. W., Solomon, H., Pike, M. C., Kroon, D., Graham, W., Abrams, M. J., Tompkins, R. G., Rubin, R. H., Barrow, S. A., and Fischman, A. J. (1993) Technetium-99m-labeled hydrazino nicotinamide derivatized chemotactic peptide analogs for imaging focal sites of bacterial infection. J. Nucl. Med. 34, 1964-1974. (11) Babich, J. W., Graham, W., Barrow, S. A., Dragotakes, S. C., Tompkins, R. H., Rubin, R. H., and Fischman, A. J. (1993) Technetium-labeled chemotactic peptides: comparison with Indium-111-labeled white blood cells for localizing acute bacterial infection in the rabbit. J. Nucl. Med. 34, 2176-2181. (12) Moyer, B. R., Vallabhajosula, S., Lister-James, J., Bush, L. R., Cyr, J. E., Snow, D. A., Bastidas, D., Lipszyc, H., and Dean, R. T. (1996) Technetium-99m-white blood cell-specific imaging agent developed from platelet factor 4 to detect infection. J. Nucl. Med. 37, 673-679. (13) Goodbody, A. E., Ballinger, J., Tran, L. L., Sumner-Smith, M., Lau, F., Meghji, K., and Pollak, A. (1994) A new Tc-99m labeled peptide inflammation imaging agent. Eur. J. Nucl. Med. 21, 790 (abstract 262). (14) Cavelier, V., Goodbody, A. E., Tran, L. L., Bossuyt, A., and Thornback, J. (1996) Human dosimetry of Tc99m-RP128, a potential inflammation imaging agent. Eur. J. Nucl. Med. 23, 1131 (abstract OMo398).

Liu et al. (15) Peers, S. H., Tran, L. L., Eriksson, S. J., Ballinger, J., and Goodbody, A. E. (1995) Imaging a model of colitis with RP128, a Tc-99m-chelated tuftsin Antagonist. J. Nucl. Med. 36, 114P (abstract 465). (16) Maina, T., Stolz, B., Albert, R., Bruns, C., Koch, P., and Ma¨cke, H. (1994) Synthesis, radiochemistry and biological evaluation of a new somatostatin analogue (SDZ 219-387) labeled with technetium-99m. Eur. J. Nucl. Med. 21, 437444. (17) Maina, T., Stolz, B., Albert, R., Nock, B., Bruns, C., and Ma¨cke, H. (1995) Synthesis, radiochemical and biological evaluation of 99mTc[N4-(D)PHE1]-octreotide, a new octreotide derivative with high affinity for somatostatin receptors. Technetium and Rhenium in Chemistry and Nuclear Medicine 4 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 395-400, SGEditorali, Padova. (18) Kolan, H., Li, J.-H., and Thakur, M. L. (1996) Sandostatin® labeled with 99mTc: In vitro stability, in vivo validity and comparison with 111In-DTPA-octreotide. Pept. Res. 9, 144150. (19) Vallabhajosula, S., Moyer, B. R., Lister-James, J., McBride, W. J., Lipszyc, H., Lee, H., Bastidas, D., and Dean, R. T. (1996) Preclinical evaluation of technetium-99m-labeled somatostatin receptor binding peptides. J. Nucl. Med. 37, 10161022. (20) Pearson, D. A., Lister-James, J., McBride, W. J., Wilson, D. M., Martel, L. J., Civitello, E. R., Taylor, J. E., Moyer, B. R., and Dean, R. T. (1996) Somatostatin receptor-binding peptides labeled with technetium-99m: chemistry and initial biological studies. J. Med. Chem. 39, 1361-1371. (21) Barrett, J. A., Heminway, S. J., Damphousse, D. J., Thomas, J. R., Looby, R. J., Edwards, D. S., Harris, T. D., Rajopadhye, M., Liu, S., and Carroll, T. R. (1994) Platelet GP IIb/IIIa antagonists in the canine arteriovenous shunt: potential thrombus imaging agents. J. Nucl. Med. 35, 52P (abstract 202). (22) Harris, T. D., Barrett, J. A., Bourque, J. P., Carroll, T. R., Damphousse, P. R., Edwards, D. S., Glowacka, D., Liu, S., Looby, R. J., Poirier, M. J., Rajopadhye, M., and Yu, K. (1994) Design and synthesis of radiolabeled GPIIb/IIIa receptor antagonists as potential thrombus imaging agents. J. Nucl. Med. 35, 245P (abstract 1005). (23) Barrett, J. A., Bresnick, M. R., Crocker, A. C., Damphousse, D. J., Hampson, J. R., Heminway, S. J., Mazaika, T. J., Kagan, M., Lazewatsky, J. L., Edwards, D. S., Liu, S., Harris, T. D., Rajopadhye, M., and Carroll, T. R. (1995) RP-431: a potential thrombus imaging agent. J. Nucl. Med. 36, 16P (abstract 55). (24) Liu, S., Edwards, D. S., Looby, R. J., Harris, A. R., Poirier, M. J., Barrett, J. A., Heminway, S. J., and Carroll, T. R. (1996) Labeling a hydrazino nicotinamide-modified cyclic IIb/IIIa receptor antagonist with 99mTc using aminocarboxylates as coligands. Bioconjugate Chem. 7, 63-71. (25) Liu, S., Edwards, D. S., Looby, R. J., Harris, A. R., Poirier, M. J., Rajopadhye, M., and Bourque, J. P. (1996) Labeling cyclic IIb/IIIa receptor antagonists with 99mTc by the preformed chelate approach: effects of chelators on properties of [99mTc]chelator-peptide conjugate. Bioconjugate Chem. 7, 196-202. (26) Barrett, J. A., Damphousse, D. J., Heminway, S. J., Liu, S., Edwards, D. S., Looby, R. J., and Carroll, T. R. (1996) Biological evaluation of 99mTc-labeled cyclic GPIIb/IIIa receptor antagonists in the canine arteriovenous shunt and deep vein thrombosis models: effects of chelators on biological properties of [99mTc]chelator-peptide conjugates. Bioconjugate Chem. 7, 203-208. (27) Edwards, D. S., Liu, S., Harris, A. R., Looby, R. J., Ziegler, M. C., Heminway, S. J., Barrett, J. A., and Carroll, T. R. (1997) A new and versatile ternary ligand system for technetium radiopharmaceuticals: water soluble phosphines and tricine as coligands in labeling a hydrazino nicotinamidemodified cyclic glycoprotein IIb/IIIa receptor antagonist with 99mTc. Bioconjugate Chem. 8, 146-154. (28) Barrett, J. A., Crocker, A. C., Damphousse, D. J., Heminway, S. J., Liu, S., Edwards, D. S., Harris, A. R., Looby, R. J., Lazewatsky, J. L., Kagan, M., Mazaika, T. J., Carroll, T. R. (1997) Biological evaluation of thrombus imaging agents

Reviews utilizing water soluble phosphines and tricine as coligands to label a hydrazinonicotinamide-modified cyclic glycoprotein IIb/IIIa receptor antagonist with 99mTc. Bioconjuate Chem. 8, 155-160. (29) Barrett, J. A., Bresnick, M., Crocker, A. C., Damphousse, D. J., Heminway, S. J., Mazaika, T. J., Kagan, M., Lazewatsky, J. L., Edwards, D. S., Liu, S., Harris, T. D., Rajpladhye, M., and Carroll, T. R. (1997) RP431: a technetium99m-labeled platelet GP IIbIIIa receptor antagonist as a thrombus imaging agent. J. Nucl. Med. (in press). (30) Knight, L. C., Radcliffe, R., Maurer, A. H., Rodwell, J. D., and Alvarez, V. L. (1994) Thrombus imaging with technetium99m synthetic peptides based upon the binding domain of a monoclonal antibody to activated platelets. J. Nucl. Med. 35, 2842-288. (31) Lister-James, J., Knight, L. C., Mauer, A. H., Bush, L. R., Moyer, B. R., and Dean, R. T. (1996) Thrombus imaging with technetium-99m-labeled, activated platelet receptor binding peptide. J. Nucl. Med. 37, 775-781. (32) Muto, P., Lastoria, S., Varrella, P., Vergara, E., Salvatore, M., Morgano, G., Lister-James, J., Bernardy, J. D., Dean, R. T., Wencker, D., and Boer, J. S. (1995) Detecting deep venous thrombosis with technetium-99m-labeled synthetic peptide P280. J. Nucl. Med. 36, 1384-1391. (33) Pearson, D. A., Lister-James, J., McBride, W. J., Wilson, D. M., Martel, L. J., Civitello, E. R., and Dean, R. T. (1996) Thrombus imaging using technetium-99m labeled high potency GPIIb/IIIa receptor antagonists. Chemistry and initial biological studies. J. Med. Chem. 39, 1372-1382. (34) Lister-James, J., Vallabhajosula, S., Moyer, B. R., Pearson, D. A., McBride, B. J., De Rosch, M. A., Bush, L. R., Machac, J., and Dean, R. T. (1997) Pre-clinical evaluation of technetium-99m platelet receptor-binding peptide. J. Nucl. Med. 38, 105-111. (35) Goldenberg, D. M. (1989) Future role of radiolabeled monoclonal antibodies in oncological diagnosis and therapy. Semin. Nucl. Med. 19, 332-339. (36) Parker, D. (1990) Tumor targeting with radiolabeled macrocycle-antibody conjugates. Chem. Soc. Rev. 19, 271291. (37) Keenan, A. M., Harbert, J. C., and Larson, S. M. (1985) Monoclonal antibodies in nuclear medicine. J. Nucl. Med. 26, 532-537. (38) Verbruggen, A. M. (1990) Radiopharmaceuticals: state of the art. Eur. J. Nucl. Med. 17, 346-364. (39) Buchsbaum, D. J., and Lawrance, T. S. (1991) Tumor therapy with radiolabeled monoclonal antibodies. Antibody, Immunoconjugates, Radiopharm. 4, 245-272. (40) Serafini, A. N. (1993) From monoclonal antibodies to peptides and molecular recognition units: an overview. J. Nucl. Med. 34, 533-536. (41) Huston, J. S., George, A. J. T., Adams, G. P., Stafford, W. F., Jamar, F., Tai, M.-S., McCartney, J. E., Oppermann, H., Heelan, B. T., Peters, A. M., Houston, L. L., Bookman, M. A., Wolf, E. J., and Weiner, L. M. (1996) Single-chain Fv radioimmunotargeting. Q. J. Nucl. Med. 40, 320-333. (42) Delaloye, A. B., and Delaloye, B. (1995) Radiolabeled monoclonal antibodies in tumor imaging and therapy: out of fashion? Eur. J. Nucl. Med. 22, 571-580. (43) Behr, T. M., and Goldenberg, D. M. (1996) Improved prospects for cancer therapy with radiolabeled antibody fragments and peptides. J. Nucl. Med. 37, 834-836 (Editorial). (44) Lister-James, J., Moyer, B. R., and Dean, R. T. (1996) Small peptides radiolabeled with 99mTc. Q. J. Nucl. Med. 40, 221233. (45) McAfee, J. G., and Neumann, R. D. (1996) Radiolabeled peptides and other ligands for receptors overexpressed in tumor cells for imaging neoplasms. Nucl. Med. Biol. 23, 673676. (46) Jones, A. G., Orvig, C., Trop, H. S., Davison, A., and Davis, M. A. (1980) A survey of reducing agents for the synthesis of tetraphenylarsonium oxotechnetiumbis(ethanedithiolate) from [99Tc]pertechnetate in aqueous solution. J. Nucl. Med. 21, 279-281.

Bioconjugate Chem., Vol. 8, No. 5, 1997 631 (47) Davison, A., Jones, A. G., Orvig, C., and Sohn, M. (1981) A new class of oxotechnetium (5+) chelate complexes containing a TcON2S2 core. Inorg. Chem. 20, 1629-1632. (48) Abrams, M. J., Davison, A., Jones, A. G., and Costello, C. E. (1983) Synthesis and characterization of new Tc(IV) cation: Tris(acetylacetonato)-technetium(IV) tetrafluoroborate. Inorg. Chim. Acta 77, L235-L236. (49) Edwards, D. S., Liu, S., Poirier, M. J., Zhang, Z., Webb, G. A., and Orvig, C. (1994) Characterization of tris(N-substituted2-methyl-3-hydroxy-4- pyridinonato)technetium(IV) cations. Inorg. Chem. 33, 5607-5609. (50) Deutsch, E., Vanderheyden, J.-L., Gerundini, P., Libson, K., Hirth, W., Colombo, F., Savi, A., and Fazio, F. (1987) Development of nonreducible technetium-99m(III) cations as myocardial perfusion imaging agents: initial experience in humans. J. Nucl. Med. 28, 1870-1880. (51) Vanderheyden, J.-L., Libson, K., Nosco, D. L., Ketering, A. R., and Deutsch, E. (1983) Preparation and characterization of [99mTc(DMPE)2X2]+, X ) Cl, Br (DMPE ) 1,2-bis(dimethylphosphino)ethane). Int. J. Appl. Radiat. Isot. 34, 1611-1618. (52) Doyle, M. N., Libson, K., Woods, M., Sullivan, J. C., and Deutsch, E. (1986) Electron transfer reactions of technetium complexes. 1. Rate of the self-exchange reaction of the Tc(I)/ Tc(II) couple [Tc(DMPE)3]+/2+, where DMPE ) 1,2-bis(dimethylphosphino)ethane). Inorg. Chem. 25, 3367-3371. (53) Abrams, M. J., Davison, A., Jones, A. G., Costello, C. E., and Pang, H. (1983) Synthesis and characterization of hexakis(alkyl isocyanide) and hexakis(aryl isocyanide) complexes of technetium (I). Inorg. Chem. 22, 2798-2800. (54) Rao, T. N., Adhikesavalu, D., Camerman, A., and Fritzberg, A. R. (1990) Technetium(V) and rhenium(V) complexes of 2,3bis(mercaptoacetamido)-propanoate. Chelate ring stereochemistry and influence on chemical and biological properties. J. Am. Chem. Soc. 112, 5798-5804. (55) Eshima, D., Taylor, A., Jr., Fritzberg, A. R., Kasina, S., Hansen, L., and Sorenson, J. F. (1987) Animal evaluation of technetium-99m triamide mercaptide complexes as potential renal imaging agents. J. Nucl. Med. 28, 1180-1186. (56) Vanbilloen, H. P., De Roo, M. J., and Verbruggen, A. M. (1996) Complexes of technetium-99m with tetrapeptides containing one alanyl and three glycyl moieties. Eur. J. Nucl. Med. 23, 40-48. (57) Subhani, M., Cleynhens, B., Bormans, G., Hoogmartens, M., De Roo, M., and Verbruggen, A. M. (1989) Complexes of technetium-99m with mercaptoacetyltripeptides: labeling characteristics and biodistribution in mice. Technetium and Rhenium in Chemistry and Nuclear Medicine 3 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 453-461, Cortina International, Verona. (58) Bormans, G., Cleynhens, B., Hoogmartens, M., De Roo, M., and Verbruggen, A. M. (1989) Synthesis and biological evaluation of L-alanyl derivatives of 99mTc-MAG3. Technetium and Rhenium in Chemistry and Nuclear Medicine 3 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 661-666, Cortina International, Verona. (59) Bormans, G., Cleynhens, B., Adriaens, P., De Roo, M., and Verbruggen, A. M. (1993) Synthesis and labeling characteristics of 99mTc-mercaptoacetyltripeptides. J. Labelled Compounds Radiopharm. 33, 1065-1078. (60) Vanbilloen, H. P., Bormans, G. M., De Roo, M. J., and Verbruggen, A. M. (1996) Complexes of technetium-99m with tetrapeptides, a new class of 99mTc-labelled agents. Nucl. Med. Biol. 22, 325-338. (61) Edwards, D. S., Cheesman, E. H., Watson, M. W., Maheu, L. J., Nguyen, S. A., Dimitre, L., Nason, T., Watson, A. D., and Walovitch, R. (1990) Synthesis and characterization of technetium and rhenium complexes of N, N′-1,2-ethylenediylbis-L-cysteine. Neurolite® and its metabolites. Technetium and Rhenium in Chemistry and Nuclear Medicine 3 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 431-444, Cortina International, Verona. (62) Oya, S., Kung, M.-P., Frederick, D., and Kung, H. F. (1995) New bisaminoethanethiol (BAT) ligands which form two interconvertable Tc-99m complexes. Nucl. Med. Biol. 22, 749757.

632 Bioconjugate Chem., Vol. 8, No. 5, 1997 (63) Kung, H. F., Guo, Y. Z., Yu, C. C., Billings, J., Subramanyam, V., and Calabrese, J. C. (1989) New brain perfusion imaging agents based on 99mTc-bis(aminoethanethiol) complexes: stereoisomers and biodistribution. J. Med. Chem. 32, 433-437. (64) Mach, R. H., Kung, H. F., Guo, Y. Z., Yu, C. C., Subramanyam, V., and Calabrese, J. C. (1989) Synthesis, characterization and biodistribution of neutral and lipid-soluble 99mTc-PAT-HM and 99mTc-TMR for brain imaging. Nucl. Med. Biol. 16, 829-837. (65) Francesconi, L. C., Graczyk, G., Wehrli, S., Shaikh, S. N., McClinton, D., Liu, S., Zubieta, J., and Kung, H. F. (1993) Synthesis and characterization of neutral MVO (M ) Tc, Re) Amine-thiol complexes containing a pendant phenylpiperidine group. Inorg. Chem. 32, 3114-3124. (66) Meegalla, S., Plu¨ssl, K., Kung, M.-P., Chumpradit, S., Stevenson, D. A., Frederick, D., and Kung, H. F. (1996) Tc99m-labeled tropanes as dopamine transporter imaging agents. Bioconjugate Chem. 7, 421-429. (67) O’Neil, J. P., Wilson, S. R., and Katzenellenbogen, J. A. (1994) Preparation and structural characterization of monoamine-monoamide bis(thiol) oxo complexes of technetium(V) and rhenium(V). Inorg. Chem. 33, 319-323. (68) Lever, S. Z., Baidoo, K. E., and Mahmood, A. (1990) Structure proof of syn/anti isomerism in N-alkylated diaminedithiol (DADT) complexes of technetium. Inorg. Chim. Acta 176, 183-184. (69) Marchi, A., Marvelli, L., Rossi, R., Magon, L., Bertolasi, V., Ferretti, V., and Gilli, P. (1992) Nitrido- and oxotechnetium(V) chelate complexes with N2S2 ligands: Synthesis and crystal structures. J. Chem. Soc., Dalton Trans., 1485-1490. (70) Spyriounis, D. M., Pelecanou, M., Stassinopoulou, C. I., Raptopoulou, C. P., Terzis, A., and Chiotellis, E. (1995) Synthesis and characterization of oxotechnetium(V) complexes with aza-substituted 2,6-dimethyl-4-azaheptane-2,6dithiol ligands and benzyl mercaptan as coligand. Inorg. Chem. 34, 1077-1082. (71) Mastrostamatis, S. G., Papadopoulos, M. S., Pirmettis, I. C., Paschali, E., Varvarigou, A. D., Stassinopoulou, C. I., Raptopoulou, C. P., Terzis, A., and Chiotellis, E. (1994) Tridentate ligands containing the SNS donor atom set as a novel backbone for the development of technetium brainimaging agents. J. Med. Chem. 37, 1077-1082. (72) Pirmettis, I. C., Papadopoulos, M. S., Mastrostamatis, S. G., Raptopoulou, C. P., Terzis, A., and Chiotellis, E. (1996) Synthesis and characterization of oxotechnetium(V) mixedligand complexes containing a tridentate N-substituted bis(2-mercaptoethyl)amine and a monodentate thiol. Inorg. Chem. 35, 1685-1691. (73) Papadopoulos, M. S., Pelecanou, M., Pirmettis, I. C., Spyriounis, D. M., Raptopoulou, C. P., Terzis, A., Stassinopoulou, C. I., and Chiotellis, E. (1996) A new donor system [(SNN)(S)] for the synthesis of oxotechnetium(V) mixed-ligand complexes. Inorg. Chem. 35, 4478-4483. (74) Klingensmith III, W. C., Fritzberg, A. R., Spitzer, V. M., Johnson, D. L., Kuni, C. C., Williamson, M. R., Washer, G., and Weil III, R. (1984) Clinical evaluation of Tc-99m N,N′bis(mercaptoacetyl)-2,3-diaminopropanoate as a replacement for I-131 hippurate: concise communication. J. Nucl. Med. 25, 42-48. (75) Walovitch, R. C., Cheesman, E. H., Maheu, L. J., and Hall, K. M. (1994) Studies of the retention mechanism of the brain perfusion imaging agent 99mTc-Bicisate (99mTc-ECD). J. Cerebral Blood Flow Metab. 14, S4-S11. (76) Harris, T. D., Edwards, D. S., and Platts, S. H. (1992) Synthesis and characteristics of isomers L,L, D,D, and D,L of Tc-99m-ECD in monkeys. J. Nucl. Med. 33, 979-980. (77) Kung, H. F., Bradshaw, J. E., Chumpradit, S., Zhang, Z. P., Kung, M. P., Mu, M., and Frederick, D. (1995) New TcO(III) and ReO(III) N2S2 complexes as potential CNS 5-HT1A receptor imaging agents. In Technetium and Rhenium in Chemistry and Nuclear Medicine 4 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 293-298, SGEditorali, Padova. (78) Meegalla, S., Plo¨ssl, K., Kung, M-P., Stevenson, D. A., Liable-Sand, L. M., Rheingold, A. L., and Kung, H. F. (1995)

Liu et al. First example of a 99mTc complex as a dopamine transporter imaging agent. J. Am. Chem. Soc. 117, 11037-11038. (79) Meegalla, S., Plo¨ssl, K., Kung, M-P., Chumpradt, S., Stevenson, D. A., Kushner, S. A., McElgin, W. T., Mozley, P. D., and Kung, H. F. (1997) Synthesis and characterization of technetium-99m-labeled tropanes as dopamine transporterimaging agnets. J. Med. Chem. 40, 9-17. (80) Treher, E. N., Francesconi, L. C., Gougoutas, J. Z., Malley, M. F., and Nunn, A. D. (1989) Monocapped tris(dioxime) complexes of technetium(III): synthesis and structural characterization of TcX(dioxime)3B-R (X ) Cl, Br; dioxime ) dimethylglyoxime, cyclohexanedione dioxime; R ) CH3, and C4H9). Inorg. Chem. 28, 3411-3416. (81) Linder, K. E., Malley, M. F., Gougoutas, J. Z., Unger, S. E., and Nunn, A. D. (1990) Neutral, seven-coordinated dioxime complexes of technetium(III): synthesis and characterization. Inorg. Chem. 29, 2428-2434. (82) Linder, K. E., Nowotnik, D. P., Malley, M. F., Gougoutas, J. Z., and Nunn, A. D. (1991) An unexpected by-product obtained during the preparation of technetium(III) boronic acid adducts of dioximes. The single crystal structure of TcCl(DMG)2(BDI)BEt (DMG ) dimethylglyoxime, BDI ) butane2,3-dione imine-oxime). Inorg. Chim. Acta 190, 249-255. (83) Marmion, M. E., Woulfe, S. R., Newmann, W. L., Pilcher, G., and Nosco, D. L. (1996) Synthesis and characterization of novel N3O3-Schiff base complexes of 99gTc, and in vivo imaging studies with analogous 99mTc complexes. Nucl. Med. Biol. 23, 567-584. (84) Linder, K. E., Wen, M. D., Nowotnik, D. P., Malley, M. F., Gougoutas, J. Z., Nunn, A. D., and Eckelman, W. C. (1991) Technetium labeling of monoclonal antibodies with functionalized BATO’s: 1. TcCl(DMG)3 PITC. Bioconjugate Chem. 2, 160-170. (85) Linder, K. E., Wen, M. D., Nowotnik, D. P., Ramalingam, K., Sharkey, R. M., Yost, F., Narra, R. K., Nunn, A. D., and Eckelman, W. C. (1991) Technetium labeling of monoclonal antibodies with functionalized BATO’s: 2. TcCl(DMG)3CPITC labeling of B72.3 and NP-4 whole antibodies and NP-4 F(ab′)2. Bioconjugate Chem. 2, 407-414. (86) Lanteingne, D., and Hnatowich, D. J. (1984) The labeling of DTPA coupled proteins. Int. J. Radiat. Isot. 35, 617-621. (87) Jurisson, S., Aston, K., Fair, C. K., Schlemper, E. O., Sharp, P. R., and Troutner, D. E. (1987) Effect of ring size on properties of technetium amine oxime complexes. X-ray structures of TcO2Pent(AO)2, which contains an unusual eight-membered chelate ring, and of TcOEn(AO)2. Inorg. Chem. 26, 3576-3582. (88) Ianoz, E., Mantegazzi, D., and Lerch, P. (1989) Preparation, crystal and molecular structure of trans-dioxo(1,4-dithia-8,11diazacyclotetradecane)- technetium(V) hexafluorophosphate. Inorg. Chim. Acta 156, 235-239. (89) Kelly, J. D., Forster, A. M., Archer, C. M., Booker, F. S., Canning, L. R., Chiu, K. W., Edwards, B., Gill, H. K., McPartlin, M., Nagle, K. R., Latham, I. A., Pickett, R. D., Storey, A. E., and Webbon, P. M. (1993) Technetium-99mtetrofosmin as a new radiopharmaceutical for myocardial perfusion imaging. J. Nucl. Med. 34, 222-227. (90) Higley, B., Smith, F. W., Gemmell, H. G., Gupta, P. D., Gvozdanovic, D. V., Graham, D., Hinge, D., Davidson, J., and Lahiri, A. (1993) Technetium-99m1,2-bis[bis(2-ethoxyethyl)phosphino]ethane: human biodistribution, dosimetry and safety of a new myocardial perfusion imaging agent. J. Nucl. Med. 34, 30-38. (91) Jones, S., and Hendel, R. C. (1993) Technetium-99m tetrofosmin: a new myocardial perfusion agent. J. Nucl. Med. Technol. 21, 191-195. (92) Schwochau, K. (1994) Technetium radiopharmaceuticalsfundamentals, synthesis, structure, and development. Angew. Chem., Int. Ed. Engl. 33, 2258-2267. (93) Abrams, M. J., Juweid, M., tenKate, C. I., Schwartz, D. A., Hauser, M. M., Gaul, F. E., Fuccello, A. J., Rubin, R. H., Strauss, H. W., and Fischman, A. J. (1990) Technetium-99mhuman polyclonal IgG radiolabeled via the hydrazino nicotinamide derivative for imaging focal sites of infection in rats. J. Nucl. Med. 31, 2022-2028. (94) Schwartz, D. A., Abrams, M. J., Hauser, M. M., Gaul, F. E., Larsen, S. K., Rauh, D., and Zubieta, J. A. (1991)

Reviews Preparation of hydrazino-modified proteins and their use for the synthesis of 99mTc-protein conjugates. Bioconjugate Chem. 2, 334-336. (95) Larson, S. K., Abrams, M. J., Higgins III, J. D., Solomon, H. F., Babich, J. W., and Fischman, A. J. (1994) Technetium complexes of tricine: useful precursor for the 99mTc labeling of hydrazino nicotinamide modified human polyclonal IgG. J. Nucl. Med. 35, 105P. (96) Larson, S. K., Solomon, H. F., Caldwell, G., and Abrams, M. J. (1995) [99mTc]tricine: a useful precursor complex for the radiolabeling of hydrazinonicotinate protein conjugates. Bioconjugate Chem. 6, 635-638. (97) Eckelman, W. C., Paik, C. H., and Steigman, J. (1989) Three approaches to radiolabeling antibodies with 99mTc. Nucl. Med. Biol. 16, 171-176. (98) Fritzberg, A. R., Berninger, R. W., Hadley, S. W., and Wester, D. W. (1988) Approaches to radiolabeling of antibodies for diagnosis and therapy of cancer. Pharm. Res. 5, 325334. (99) Otsuka, F. L., and Welch, M. J. (1987) Methods to label monoclonal antibodies for use in tumor imaging. Nucl. Med. Biol. 14, 243-249. (100) Hnatowich, D. J. (1990) Antibody radiolabeling, problems and promises. Nucl. Med. Biol. 17, 49-55. (101) Hnatowich, D. J. (1990) Recent developments in radiolabeling of antibodies with iodine, indium, and technetium. Semin. Nucl. Med. 20, 80-91. (102) Srivastava, S. C., and Mease, R. C. (1991) Progress in research on ligands, nuclides and techniques for labeling monoclonal antibodies. Nucl. Med. Biol. 18, 589-603. (103) Delmon-moingeon, L. I., Mahmood, A., Davison, A., and Jones, A. G. (1991) Strategies for labeling monoclonal antibodies and antibody-like molecules with technetium-99m. J. Nucl. Biol. Med. 35, 47-59. (104) Rhodes, B. A. (1991) Direct labeling of proteins with 99mTc. Nucl. Med. Biol. 18, 667-676. (105) Eckelman, W. C., and Steigman, J. (1991) Direct labeling with 99mTc. Nucl. Med. Biol. 18, 3-7. (106) Liu, Y.-F., and Wu, C.-C. (1991) Radiolabeling of monoclonal antibodies with metal chelates. Pure Appl. Chem. 63, 427-463. (107) Griffiths, G. L., Goldenberg, D. M., Jones, A. L., and Hansen, H. J. (1992) Radiolabeling of monoclonal antibodies and fragments with technetium and rhenium. Bioconjugate Chem. 3, 91-99. (108) Bhargava, K. K., and Acharya, S. A. (1989) Labeling of monoclonal antibodies with radionuclides. Semin. Nucl. Med. 19, 187-201. (109) Wolf, W., and Shani, J. (1986) Criteria for the selection of the most desirable radionuclide for radiolabeling monoclonal antibodies. Nucl. Med. Biol. 13, 319-324. (110) Gansow, O. A. (1991) Newer approaches to the radiolabeling of monoclonal antibodies by use of metal chelates. Nucl. Med. Biol. 18, 269-281. (111) Zamora, P. O., and Rhodes, B. A. (1992) Imidazoles as well as thiolates in proteins bind technetium-99m. Bioconjugate Chem. 3, 493-498. (112) Fritzberg, A. R., Abrams, P. G., Beaumier, P. L., Kasina, S., Morgan, A. C., Rao, T. N., Reno, J. M., Sanderson, J. A., Srinivasan, A., and Wilbur, D. S. (1988) Specific and stable labeling of antibodies with technetium-99m with a diamide dithiolate chelating agent. Proc. Natl. Acad. Sci. U.S.A. 85, 4025-4029. (113) Eary, J. F., Schroff, R. W., Abrams, P. G., Fritzberg, A. R., Morgan, A. C., Kasina, S., Reno, J. M., Srinivasan, A., Woodhouse, C. S., Wilbur, D. S., Natale, R. B., Collins, C., Stehlin, J. S., Mitchell, M., and Nelp, W. B. (1989) Successful imaging of malignant melanoma with technetium-99mlabeled monoclonal antibodies. J. Nucl. Med. 30, 25-32. (114) Kasina, S., Rao, T. N., Srinivasan, A., Sanderson, J. A., Fitzner, J. N., Reno, J. M., Beaumier, P. L., and Fritzberg, A. R. (1991) Development and biological evaluation of a kit for preformed chelate technetium-99m radiolabeling of an antibody Fab fragment using a diamide dimercaptide chelating agent. J. Nucl. Med. 32, 1445-1451. (115) Majocha, R. E., Reno, J. M., Friedland, R. P., VanHaight, C., Lyle, L. R., and Marotta, C. A. (1992) Development of a

Bioconjugate Chem., Vol. 8, No. 5, 1997 633 monoclonal antibody specific for β/A4 amyloid in Alzheimer’s disease brain for application to in vivo imaging of amyloid angiopathy. J. Nucl. Med. 33, 2184-2189. (116) Liu, S., and Edwards, D. S. (1995) New N2S2 diamidedithiol and N3S triamidethiols as bifunctional chelating agents for labeling small peptides with technetium-99m. In Technetium and Rhenium in Chemistry and Nuclear Medicine 4 (M. Nicolini, G. Banoli, and U. Mazzi, Eds.) pp 383-393, SGEditorali, Padova. (117) Rajopadhye, M., Edwards, D. S., Bourque, J. P., and Carroll, T. R. (1996) Synthesis and technetium-99m labeling of cyclic GPIIb/IIIa receptor antagonists conjugated to 4,5bis(mercaptoacetylamido)pentanoic acid (mapt). Bioorg. Med. Chem. Lett. 6, 1737-1740. (118) Baidoo, K. E., and Lever, S. Z. (1990) Synthesis of a diaminedithiol bifunctional chelating agent for incorporation of technetium-99m into biomolecules. Bioconjugate Chem. 1, 132-137. (119) Lever, S. Z., Baidoo, K. E., Mahmood, A., Matsumura, K., Scheffel, U., and Wagner, H. N., Jr. (1994) Novel technetium ligand with affinity for muscarinic cholinergic receptor. Nucl. Med. Biol. 21, 157-164. (120) Lever, S. Z., Sun, S-Y., Scheffel, U. A., Kaltovich, F. A., Baidoo, K. E., Goldfarb, H., and Wagner, H. N., Jr. (1994) Pulmonary accumulation of neutral diamine dithiol complexes of technetium-99m. J. Pharm. Sci. 84, 802-809. (121) Del Rosario, R. B., Jung, Y.-W., Baidoo, K. E., Lever, S. Z., and Wieland, D. M. (1994) Synthesis and in vivo evaluation of a 99m/99Tc-DADT-Benzovesamicol: a potential marker for cholinergic neurons. Nucl. Med. Biol. 21, 197-203. (122) O’Neil, J. P., Anderson, C. J., Carlson, K. E., Welch, M. J., and Katzenellenbogen, J. A. (1993) An improved progestintechnetium complex as a potential imaging agent for steroid receptors. J. Nucl. Med. 33, 18P (abstract 61). (123) Eisenhut, M., Miβfeldt, M., Lehmann, W. D., and Karas, M. (1991) Synthesis of a bis(aminoethanethiol) ligand with an activated ester group for protein conjugation and 99mTclabeling. J. Labelled Comp. Radiopharm. 29, 1283-1291. (124) Eisenhut, M., Lehmann, W. D., Becker, W., Elser, H., Strittmatter, W., Baum, R. P., Valerius, T., Repp, R., and Deo, Y. (1996) Bifunctional NHS-BAT ester for antibody conjugation and stable technetium-99m labeling: conjugation chemistry, immunoreactivity and kit formulation. J. Nucl. Med. 37, 362-370. (125) DiZio, J. P., Fiashi, R., Davison, A., Jones, A. G., and Katzenellenbogen, J. A. (1991) Progestin-rhenium complexes: metal labeled steroids with high receptor binding affinity, potential receptor-directed agents for diagnostic imaging or therapy. Bioconjugate Chem. 2, 352-366. (126) O’Neil, J. P., Carlson, K. E., Anderson, C. J., Welch, M. J., and Katzenellenbogen, J. A. (1994) Progestin radiopharmaceuticals labeled with technetium and rhenium: synthesis, binding affinity, and in vivo distribution of a new progestin N2S2-metal conjugate. Bioconjugate Chem. 5, 182-193. (127) DiZio, J. P., Anderson, C. J., Davison, A., Ehrhardt, G. J., Carlson, K. E., Welch, M. J., and Katzenellenbogen, J. A. (1992) Technetium- and rhenium-labeled progestins: synthesis, receptor binding and in vivo distribution of an β-substituted progestin labeled with technetium-99m and rhenium-186. J. Nucl. Med. 33, 558-569. (128) Liu, S., Edwards, D. S., Harris, A. R., and Singh, P. R. (1997) 99mTc-labeling kinetics of four thiol-containing chelators and 2-hydrazinopyridine: factors influencing their radiolabeling efficiency. Appl. Radiat. Isot. (in press). (129) Bryson, N., Lister-James, J., Jones, A. G., Davis, W. M., and Davison, A. (1990) Protecting groups in the preparation of thiolate complexes of technetium. Inorg. Chem. 29, 29482951. (130) Bryson, N., Dewan, J. C., Lister-James, J., Jones, A. G., and Davison, A. (1988) Neutral technetium(V) complexes with amide-thiol-thioether chelate ligands. Inorg. Chem. 27, 21542161. (131) Anderson, F. A., Wheeler, H. B., Goldberg, R. T., Hosner, D. W., Patwardhar, N. A., Jovanovic, B., Forcier, A., and Dalen, J. E. A. (1991) A population-based perspective of the

634 Bioconjugate Chem., Vol. 8, No. 5, 1997 hospital incidence and case facility rates of deep vein thrombosis and pulmonary embolism. Ann. Intern. Med. 151, 933938. (132) Hull, R. D., Hirsh, J., Carter, D. J., Jay, R. M., Dodd, P. E., Ockelford, P. A., Coates, G., Gill, G. J., Turpie, A. G., Dayle, D. J., Buller, H. R., and Raskob, G. E. (1983) Pulmonary angiography, ventilation lung scanning and venography for clinically suspected pulmonary embolism with abnormal perfusion lung scan. Ann. Intern. Med. 98, 891938. (133) Knight, L. C. (1990) Radiopharmaceuticals for thrombus detection. Semin. Nucl. Med. 20, 52-67. (134) Knight, L. C. (1993) Scintigraphic methods for detecting vascular thrombus. J. Nucl. Med. 34, 554-561. (135) Wasser, M. N. J. M., and Pauwels, E. K. J. (1990) Immunoscintigraphy of thrombosis. Eur. J. Nucl. Med. 16, 583-585 (Editorial). (136) Loscalzo, J., and Rocco, T. P. (1992) Imaging arterial thrombi: an elusive goal. Circulation 85, 382-384. (137) Som, P., and Oster, Z. H. (1994) Thrombus-specific imaging: approaching the elusive goal. J. Nucl. Med. 35, 202-203 (Editorial). (138) Flank, C., Kakkar, V. V., and Clarke, M. B. (1968) The detection of venous thrombosis of the legs using 125I-labelled fibrinogen. Br. J. Surg. 55, 742-747. (139) Negus, D., Pinto, D. J., LeQuensne, L. P., Brown, N., and Chapman, M. (1968) 125I-labelled fibrinogen in diagnosis of deep-vein thrombosis and its correlation with phlebography. Br. J. Surg. 55, 835-839. (140) Thakur, M. L., Welch, M. J., Joist, J. H., and Coleman, R. E. (1976) Indium-111-labeled platelets: studies on preparation and evaluation of in vitro and in vivo functions. Thromb. Res. 9, 345-357. (141) Ezekowitz, M. D., Leonard, J. C., Smith, E. O., Allen, E. W., and Taylor, F. B. (1981) Identification of left ventricular thrombi in man using indium-111-labeled autologous platelets. Circulation 63, 801-810. (142) Stratton, J. R., Ritchie, J. L., Hamilton, G. W., Hammermeister, K. E., and Harker, L. A. (1981) Left ventricular thrombi: in vivo detection by indium-111 platelet imaging and two dimentional echocardiography. Am. J. Cardiol. 47, 874-881. (143) Ezekowitz, M. D., Eichner, E. R., Scatterday, R., and Elkins, C. R. (1982) Diagnosis of a persistent pulmonary embolus by indium-111 platelet scintigraphy with angiography and tissue confirmation. Am. J. Med. 72, 839-842. (144) Davis, H. H., II, Siegel, B. A., Sherman, L. A., Heaton, W. A., and Welch, M. J. (1980) Scintigraphy with 111In-labeled autologous platelets in venous thromboembolism. Radiology 136, 203-207. (145) Fenech, A., Hussey, J. K., Smith, F. W., Dendy, P. P., Bennett, B., and Douglas, A. S. (1981) Diagnosis of deep vein thrombosis using autologous indium-111-labeled platelets. Br. Med. J. 282, 1020-1022. (146) Stratton, J. R. (1991) Thrombosis imaging with indium111-labeled platelets. Cardiac ImagingsPrinciples and Practice (M. L. Marcus, H. R. Schelbert, D. J. Skorton, and D. J. Wolf, Eds.) pp 1121-1134, W. B. Saunders, New York. (147) Stratton, J. R., Cerqueira, M. D., Dewhurst, T. A., and Kohler, T. R. (1994) Imaging arterial thrombosis: comparison of technetium-99m-labeled monoclonal antifibrin antibodies and indium-111-platelets. J. Nucl. Med. 35, 1731-1737. (148) Ali, F. E., Bennett, D. B., Calvo, R. R., Elliott, J. D., Hwang, S. M., Ku, T. W., Lago, M. A., Nichols, A. J., Romoff, T. T., Shah, D. H., Vasko, J. A., Wong, A. S., Yellin, T. O., Yuan, C-K., and Samanen, J. M. (1994) Conformationally constrained peptides and semipeptides derived from RGD as potent inhibitors of the platelet fibrinogen receptor and platelet aggregation. J. Med. Chem. 37, 769-780, and references cited therein. (149) Cheng, S., Craig, W. S., Mullen, D., Tschopp, J. F., Dixon, D., and Pierschbacher, M. D. (1994) Design and synthesis of novel cyclic RGD- containing peptides as highly potent and selective integrin RIIbβ3 antagonists. J. Med. Chem. 37, 1-8, and references cited therein.

Liu et al. (150) Teng, W., Rose, J. W., Phillips, D. R., Nannizzi, L., Arsen, A., Campbell, A. M., and Charo, I. F. (1993) Design of potent and specific integrin antagonists. J. Biol. Chem. 268, 10661073. (151) Zablocki, J. A., Miyano, M., Garland, R. B., Pireh, D., Schretzman, L., Rao, S. N., Lindmark, R. J., Panzer-Knodle, S. G., Nicholson, N. S., Taite, B. B., Salyers, A. K., King, L. W., Campion, J. G., and Feigen, L. P. (1993) Potent in vitro and in vivo inhibitors of platelet aggregation based upon the Arg-Gly-Asp-Phe sequence of fibrinogen. A proposal on the nature of the binding interaction between the Arg-guanidine of RGDX mimetics and the platelet GP IIb/IIIa receptor. J. Med. Chem. 36, 1811-1919. (152) Bach, A. C., II, Eyermann, C. J., Gross, J. D., Bower, M. J., Harlow, R. L., Weber, P. C., and DeGrado, W. F. (1994) Structural studies of a family of high affinity ligands for GPIIb/IIIa. J. Am. Chem. Soc. 116, 3207-3219. (153) Jackson, S., DeGrado, W. F., Dwivedi, A., Parthasarathy, A., Higley, A., Krywko, J., Rockwell, A., Markwalder, J., Wells, G., Wexler, R., Mousa, S., and Harlow, R. L. (1994) Template-constrained cyclic peptides: design of high-affinity ligands for GPIIb/IIIa. J. Am. Chem. Soc. 116, 3220-3230. (154) Xue, C.-B., and DeGrado, W. F. (1995) An efficient synthesis of glycoprotein IIb/IIIa inhibitor DMP728. A novel synthesis of NR-methylarginine-containing peptide. J. Org. Chem. 60, 946-952. (155) Wityak, J., Fevig, J. M., Jackson, S. A., Johnson, A. L., Mousa, S. A., Parthasarthy, A., Wells, G. J., DeGrado, W. F., and Wexler, R. R. (1995) Synthesis and antiplatelet activity of DMP757 analogs. Bioorg. Med. Chem. Lett. 5, 2097-2100. (156) Harris, T. D., Rajopadhye, M., Damphousse, P. R., Glowacka, D., Yu, K., Bourque, J. P., Barrett, J. A., Damphousse, D. J., Heminway, S. J., Lazewatsky, J. L., Mazaika, T., and Carroll, T. R. (1996) Tc-99m-labeled fibrinogen receptor antagonists: design and synthesis of cyclic RGD peptides for the detection of thrombi. Bioorg. Med. Chem. Lett. 6, 1741-1746. (157) Rajopadhye, M., Harris, T. D., Yu, K., Glowacka, D., Damphousse, P. R., Barrett, J. A., Heminway, S. J., Edwards, D. S., and Carroll, T. R. (1997) Synthesis, evaluation and Tc99m complexation of a hydrazinonicotinyl conjugate of a GP IIb/IIIa antagonist cyclic peptide for the detection of deep vein thrombosis. Bioorg. Med. Chem. Lett. 7, 955-960. (158) Liu, S., and Edwards, D. S. Unpublished results. (159) Abrams, M. J., Larsen, S. K., Shaikh, S. N., and Zubieta, J. (1991) Investigation of technetium-organohydrazine coordination chemistry. The crystal and molecular structures of [TcCl2(C8H5N4)(PPh3)2]‚0.75C7H8 and [TcNCl2(PPh3)2]‚0.25CH2Cl2. Inorg. Chim. Acta 185, 7-15. (160) Archer, C. M., Dilworth, J. R., Jobanputra, P., Thompson, R. M., McPartlin, M., Povey, D. C., Smith, G. W., and Kelly, J. D. (1990) Development of new technetium cores containing technetium-nitrogen multiple bonds. Synthesis and characterization of some diazenido-, hydrazido- and imido- complexes of technetium. Polyhedron 9, 1497-1502. (161) Dilworth, J. R., Jobanputra, P., Thompson, R. M., Archer, C. M., Povey, D. C., Kelly, J. D., and Hiller, W. (1992) Crystal structure of a diazenido- dithiocarbamate complex of technetium, [Tc(NNC6H4Cl)((CH3)2NCS2)2(PPh3)]. Z. Naturforsch. 46, 449-452. (162) Archer, C. M., Dilworth, J. R., Jobanputra, P., Thompson, R. M., McPartin, M., and Hiller, W. (1993) Technetium diazenido complexes. Part 1. Synthesis and structures of [TcCl(NNC6H4Cl-4)2(PPh3)2] and [TcCl(NNPh)(Ph2PCH2CH2PPh2)2][PF6]‚H2O. J. Chem. Soc., Dalton Trans., 897-904. (163) Dilworth, J. R., Jobanputra, P., Thompson, R. M., Povey, D. C., Archer, C. M., and Kelly, J. D. (1994) Technetium diazenido complexes. Part 2. Substitution chemistry of structures of [TcCl(NNC6H4Cl-4)2(PPh3)2] and the synthesis of technetium diazenido-complexes directly from [NH4][TcO4]. J. Chem. Soc., Dalton Trans., 1251-1256. (164) Nicholson, T., de Vries, N., Davison, A., and Jones, A. G. (1989) Synthesis and characterization of aryldiazenido technetium complexes and their protonation reactions. The X-ray structure of [TcCl(PPh3)2(NNC6H4Br)2]. In Technetium and Rhenium in Chemistry and Nuclear Medicine 3 (M. Nicolini,

Reviews G. Bandoli, and U. Mazzi, Eds.) pp 95-108, Cortina International, Verona. (165) Cook, J., Davision, A., Jones, A. J., and Davis, W. M. (1990) The reaction chemistry of HTc(CO)3(PPh3)2. In Technetium and Rhenium in Chemistry and Nuclear Medicine 3 (M. Nicolini, G. Bandoli, and U. Mazzi, Eds.) pp 65-68, Cortina International, Verona. (166) Nicholson, T., Cook, J., Davison, A., Rose, D. J., Maresca, K. P., Zubieta, J. A., and Jones, A. J. (1996) The synthesis and characterization of [MCl3(NdNC5H4NH)(HNdNC5H4N)] from [MO4]- (where M ) Re, Tc) organodiazenido, organodiazene-chelate complexes. The X-ray structure of [ReCl3(NdNC5H4NH)(HNdNC5H4N)]. Inorg. Chim. Acta 252, 421426. (167) Nicholson, T., Cook, J., Davison, A., Rose, D. J., Maresca, K. P., Zubieta, J. A., and Jones, A. J. (1996) The synthesis, characterization and X-ray crystal structure of the rhenium organodiazenido, organodiazene complex of [ReCl2(PPh3)(NdNC5H4N)(HNdNC5H4N)]. Inorg. Chim. Acta 252, 427430. (168) Oyen, W. J. G., Boerman, O. C., van der Laken, C. J., Claessens, R. A. M. J., van der Meer, J. W. M., and Corsten, F. H. M. (1996) The uptake mechanisms of inflammation- and infection-localizing agents. Eur. J. Nucl. Med. 23, 459-465. (169) Boxen, I., and Ballinger, J. R. (1991) Nuclear Medicine detection of inflammation and infection. Curr. Opin. Radiol. 3, 840-850. (170) McAfee, J. G. (1990) What is the best method for imaging focal infections? J. Nucl. Med. 31, 413-416 (Editorial). (171) Claessens, R. A. M. J., Koenders, E. B., Oyen, W. J. G., and Corstens, F. H. M. (1996) Retention of technetium-99m in infectious foci in rats after release from technetiun-99m labelled human non-specific polyclonal immunoglobulin G: a dual-label study with hydrazinonicotinamido and iminothiolano immunoglobulin. Eur. J. Nucl. Med. 23, 1536-1539. (172) Buscombe, J. R., Lui, D., Ensing, G., de Jong R., and Ell, P. J. (1990) 99mTc-human immunoglobulin (HIG)-first results of a new agent for the localization of infection and inflammation. Eur. J. Nucl. Med. 16, 649-655. (173) Lei, K., Rusckowski, M., Chang, F., Qu, T., Mardirossion, G., and Hnatowich, D. J. (1996) Technetium-99m antibodies labeled with MAG3 and SHNH: an in vitro and animal in vivo comparison. Nucl. Med. Biol. 23, 917-922. (174) Barrow, S. A., Graham, W., Jyawook, S., Dragotakes, S. C., Solomon, H. F., Babich, J. W., Rubin, R. H., and Fischman, A. J. (1993) Localization of indium-111-immunoglobulin G, technetium-99m-immunoglobulin G and indium-111-labeled white blood cells at sites of acute bacterial infection in rabbits. J. Nucl. Med. 34, 1975-1979. (175) Callahan, R. J., Barrow, S. A., Abrams, M. J., Rubin, R. H., and Fischman, A. J. (1996) Biodistribution and dosimetry of technetium-99m-hydrazino nicotinamide IgG: comparison with indium-111-DTPA-IgG. J. Nucl. Med. 37, 843-846. (176) Fischman, A. J., Solomon, H. F., Babich, J. W., Abrams, M. J., Callahan, R. J., and Strauss, H. W. (1994) Imaging of focal sites of inflammation in rhesus monkeys with 99mTclabeled human polyclonal IgG. Nucl. Med. Biol. 21, 111-116 (177) Peters, A. M., Danpure, H. J., Osman, S., Halker, R. J., Henderson, B. L., Hodgson, H. J., Kelly, J. D., Neirinckx, R. D., and Lavender, J. P. (1986) Clinical experience with 99mTchexamethylpropyleneamineoxime for labeling leukocytes and imaging inflammation. Lancet 2, 946-949. (178) Vorne, M., Soini, I., Lantto, T., and Paakkinen, S. (1989) Technetium-99m-HM-PAO-labeled leukocytes in detection of inflammatory lesions: comparison with gallium-67 citrate. J. Nucl. Med. 30, 1332-1336. (179) Charron, M., Orenstein, S. R., and Bhargava, S. (1994) Detection of inflammatory bowel disease in pediatric patients with technetium-99m-HMPAO-labeled leukocytes. J. Nucl. Med. 35, 451-455. (180) Peters, A. M. (1996) The choice of an appropriate agent for imaging inflammation. Nucl. Med. Commun. 17, 455-458. (181) Lange, J. M. A., Boucher, C. A. B., Hollak, C. E. M., Wiltink, E. H. H., Reiss, P., van Royen, E. A., Roos, M., Danner, S. D., and Goudsmit, J. (1990) Failure of zidovudine prophylaxis after accidental exposure to HIV-1. N. Engl. J. Med. 322, 1375-1377.

Bioconjugate Chem., Vol. 8, No. 5, 1997 635 (182) Rojas-Burke, J. (1990) Health officials reacting to infection mishaps. J. Nucl. Med. 33, 13-27. (183) Edwards, D. S., Barrett, J. A., Liu, S., Ziegler, M. C., Mazaika, T., Vining, M., Bridger, G., Higgins, III, J., and Abrams, M. J. (1996) A stabilized Tc-99m complex of a chemotactic peptide-HYNIC conjugate for imaging infection. Eur. J. Nucl. Med. 23, 1142 (abstract Omo440). (184) Pollak, A., Goodbody, A. E., Ballinger, J. R., Duncan, G. S., Tran, L. L., Dunn-Dufault, R., Meghji, K., Lau, F., Andrey, T. W., Boxen, I., and Sumner-Smith, M. (1996) Imaging inflammation with 99mTc-labelled chemotactic peptides: analogues with reduced neutropenia. Nucl. Med. Commun. 17, 132-139. (185) Solomon, H. F., Derlan, C. K., Beblavy, M., Jester, D., Santull, R., Pike, M., Kroon, D., Hoey, K., and Fischman, A. J. (1994) Focal infection imaging using an In- 111 labeled antagonist chemotactic peptide. J. Nucl. Med. 35, 45P (abstract 172). (186) Reubi, J. C. (1993) The role of peptides and their receptors as tumor markers. Endocrinol. Metab. Clin. North Am. 22, 917-939. (187) Virgolini, I., Pangerl, T., Bischof, C., Leimer, M., Kartaran, A., Yang, Q., Peck-Radosavjlevic, M., Kaserer, K., Niederle, B., Angelberger, P., Gangl, A., and Valent, P. (1996) Somatostatin (SST) and vasoactive intestinal peptide (VIP) receptor® subtype expression. Eur. J. Nucl. Med. 23, 1101 (abstract Omo274). (188) Maini, C. L., Tofani, A., Scuito, R., Carapella, C., Cioffi, R., and Crecco, M. (1993) Somatostatin receptors in menigeomas: a scintigraphic study using 111In- DTPA-Phe-1octreotide. Nucl. Med. Commun. 14, 550-558. (189) Reubi, J. C. (1995) Neuropeptide receptors in health and disease: the molecular basis for in vivo imaging. J. Nucl. Med. 36, 1825-1835. (190) Bardfeld, P. A., Chervu, L. R., and Myrty, D. R. K. (1976) The organ distribution of 131I-tyrosyl somatostatin. Br. J. Radiol. 49, 381-382. (191) Wangberg, B., Nilsson, O., Theodørsson, E., Dahlstrøm, A., and Ahlman, H. (1991) The effect of a somatostatin analog on the release of hormones from human midgut carcinoid tumor cells. Br. J. Cancer 64, 23-28. (192) Lamberts, S. W. J. (1988) The role of somatostatin in the regulation of anterior pituitary hormone secretion and the use of its analogues in the treatment of human pituitary tumors. Endocrinol. Rev. 9, 417-436. (193) Bauer, W., Briner, U., Doepfner, W., Haller, R., Huguenin, R., Marbach, P., Petcher, T. J., and Pless, J. (1982) SMS 201995: a very potent and selective octapeptide analogue of somatostatin with prolonged action. Life Sci. 31, 1133-1140. (194) Weckbecker, G., Liu, R., Tolcsvai, L., and Bruns, C. (1992) Antiproliferative effects of octreotide (SMS 201-995) on ZR75-1 human breast cancer cells in vivo and in vitro. Cancer Res. 52, 4973-4978. (195) Bakker, W. H., Krenning, E. P., Breeman, W. A. P, Koper, J. W., Kooij, P. P. M., Reubi, J. C., Klijn, J. G., Visser, T. J., Docter, R., and Lamberts, S. W. J. (1990) Receptor scintigraphy with a radioiodinated somatostatin analogue: radiolabeling, purification, biologic activity and in vivo application in animals. J. Nucl. Med. 31, 1501-1509. (196) Krenning, E. P., Breeman, W. A., Bakker, W. H., Koper, J. W., Kooij, P. P., Ansema, L., Lamberts, S. W. J., and Reubi, J. C. (1989) Localization of endocrine-related tumors with radioiodinated analogue of somatostatin. Lancet 1, 242-243. (197) Bakker, W. H., Krenning, E. P., Reubi, J. C., Breeman, W. A. P., SelyonoHan, B., de Jong, M., Kooij, P. P. M., Bruns, C., van Hagen, P. M., Marbach, P., Visser, T. J., Pless, J., and Lamberts, S. W. J. (1991) In vivo application of [111InDTPA-DPhe1]-octreotide for detection of somatostatin receptor-positive tumors in rats. Life Sci. 49, 1593-1601. (198) Lipp, R. W., Silly, H., Ranner, G., Dobnig, H., Passath, A., Leb, G., and Krejs, G. J. (1995) Radiolabeled octreotide for the demonstration of somatostatin receptors in malignant lymphoma and lymphadenopathy. J. Nucl. Med. 36, 13-18. (199) Moretti, J. L., Caglar, M., Boaziz, C., Caillat-Vigneron, N., and Morere, J. F. (1995) Sequential functional imaging with technetium-99m hexakis-2-methoxy-isobutylisonitrile and indium-111 octreotide: can we predict the response to

636 Bioconjugate Chem., Vol. 8, No. 5, 1997 chemotherapy in small cell lung cancer? Eur. J. Nucl. Med. 22, 177-180. (200) Tenebaum, F., Lumbroso, J., Schlumberger, M., Mure, A., Plouin, P. F., Cailou, B., and Parmentier, C. (1995) Comparison of radiolabeled octreotide and meta-iodobenzylguanidine (MIBG) scintigraphy in malignant pheochromocytoma. J. Nucl. Med. 36, 1-6. (201) Haldemann, A. R., Ro¨sler, H., Barth, A., Waser, B., Geiger, L., Godoy, N., Markwalder, R. V., Seiler, R. W., Sutzer, M., and Reubi, J. C. (1995) Somatostatin receptor scintigraphy in central nervous system tumors: role of bloodbrain barrier permeability. J. Nucl. Med. 36, 403-410. (202) Maini, C. L., Cioffi, R. P., Tofani, A., Sciuto, R., Fontana, M., and Carapella, C. M. (1995) Indium-111 octreotide scintigraphy in neurofibromatosis. Eur. J. Nucl. Med. 22, 201-206. (203) Duet, M., Mundler, O., Ajzenberg, C. A., Berolatti, B., Chedin, P., Duranteau, L., and Warnet, A. (1994) Somatostatin receptor imaging in non-functioning pituitary adenomas: value of an uptake index. Eur. J. Nucl. Med. 21, 647-650. (204) Breeman, W. A. P., Hofland, L. J., van der Pluijm, M., van Koetsveld, P. M., de Jong, M., Setyono-Han, B., Bakker, W. H., Kwekkeboom, D. J., Visser, T. J., Lamberts, S. W. J., and Krenning, E. P. (1994) A new radiolabeled somatostatin analogue [111In-DTPA-DPhe1]RC-160: preparation, biological activity, receptor scintigraphy in rats and comparison with [111In-DTPA-DPhe1]octreotide. Eur. J. Nucl. Med. 21, 328335. (205) Ma¨cke, H. R., and Behe, M. (1996) New octreotide derivatives labelled with technetium-99m. J. Nucl. Med. 37, 29P (abstract 107).

Liu et al. (206) Behe, M., and Ma¨cke, H. R. (1995) New somatostatin analogues labelled with technetium-99m. Eur. J. Nucl. Med. 22, 791 (abstract 267). (207) Thakur, M. L., John, E., Li, J., Reddy, H. R., Halmos, G., and Schally, A. V. (1995) Tc-99m-RC-160: a somatostatin analog for imaging prostate cancerscomparison with I-125RC-160, and In-111-octreotide. J. Nucl. Med. 36, 92P (abstract 374). (208) Thakur, M. L., Halmos, G., and Jan, J. (1994) Technetium-99m-RC-160: a somatostatin analog. J. Nucl. Med. 35, 259P (abstract 1063). (209) Breeman, W. A. P., VanHagen, M. P., Visser-Wisselaar, H. A., van der Pluijm, M. E., Koper, J. W., Setyono-Han, B., Bakker, W. H., Kwekkeboom, D. J., Hazenberg, M. P., Lamberts, S. W. J., Visser, T. J., and Krenning, E. P. (1996) In vitro and in vivo studies of substance P receptor expression in rats with the new analog [Indium-111-DTPA-Arg1]substance P. J. Nucl. Med. 37, 108-117. (210) Virgolini, I., Raderer, M., Kurtaran, A., Angelberger, P., Yang, Q, Radosavljevic, M., Leimer, M., Kaserer, K., Li, S. R., Kornek, G., Hu¨bsch, Niederle, B., Pidlich, J., Scheithauer, W., and Valent, P. (1996) 123I-vasoactive intestinal peptide (VIP) receptor scanning: update of imaging results in patients with adenocarcinomas and endocrine tumors of the gastrointestinal tract. Nucl. Med. Biol. 23, 685-692. (211) Jurisson, S., Berning, D., Jia, W., and Ma, D.-S. (1993) Coordination compounds in nuclear medicine. Chem. Rev. 93, 1137-1156.

BC970058B