Home Biocomplexes in radiochemistry
Article Publicly Available

Biocomplexes in radiochemistry

  • Kazuma Ogawa EMAIL logo
Published/Copyright: May 31, 2016
Become an author with De Gruyter Brill

1 Introduction

Compounds are labeled with radionuclides and are used in nuclear medicine. These compounds are generally injected intravenously, after which they accumulate in target tissues, decay, and emit radiation. If the radiation produced is in the form of very high frequency electromagnetic waves, such as gamma rays or X-rays, it is highly penetrating and can be detected in patients by gamma scintigraphy, single photon emission computed tomography (SPECT), or positron emission tomography (PET). Therefore, images can be obtained that show the distribution of the radiopharmaceuticals within the body. The purpose of the images acquired in nuclear medicine is to provide functional information as opposed to anatomical information provided by other techniques such as X-ray, computed tomography (CT), and magnetic resonance imaging (MRI). Although the spatial resolutions of SPECT and PET are much lower than those of CT and MRI, SPECT and PET still provide useful information, such as imaging of processes at the molecular and cellular level and quantitative data useful in clinical assessments.

If a radionuclide emits radiation which has relatively low penetration and loses its high energy within a short distance, such as beta and alpha radiation, it is suitable for use in internal radionuclide therapy for cancer. The best known internal radionuclide therapy is radioimmunotherapy. In this type of therapy, antibodies to the molecules which are over expressed on the surface of cancer cells are used as carriers of radionuclides for delivery to the tumours. For this purpose, radiolabeled antibodies are injected into cancer patients. In clinical use, two successful radiolabeled antibodies, 90Y-ibritumomab tiuxetan (Zevalin®) and 131I-tositumomab (Bexxar®), which are beta particle-emitter-labeled anti-CD20 monoclonal antibodies, have been approved by the US Food and Drug Administration (FDA) for the treatment of non-Hodgkin lymphoma.

In this chapter, I present some radiometal complexes used for diagnosis and therapy and discuss my recent research, focusing on radiolabeled compounds used for bone disorders and apoptosis.

2 Bone-seeking complexes

2.1 Diagnostic bone-seeking radiopharmaceuticals

Bisphosphonates are synthetic pyrophosphate analogs that are stable in vivo because of their P-C-P central structure, rather than the P-O-P configuration of pyrophosphates, which affords greater resistance to phosphatase hydrolysis (Fig. 1a). Because bisphosphonates inhibit osteoclast-mediated bone resorption and bone turnover, they have been used in the treatment of skeletal disorders, such as osteoporosis, metastatic bone cancer, and Paget’s disease [1, 2]. Bisphosphonates have a high affinity to bones, especially to hydroxyapatite, which is a mineral present in bones. Bisphosphonates also have been used as carriers of radioisotopes to bones. For many years, two 99mTc-bisphosphonate complexes, 99mTc-methylenediphosphonate (99mTc-MDP, Fig. 1b) and 99mTc-hydroxymethylenediphosphonate (99mTc-HMDP, Fig. 1c), have been clinically used in nuclear medicine for diagnosis of bone disorders, such as metastatic bone cancer [35], because their high sensitivity can detect bone disorders before the occurrence of anatomical changes. Bone metastases are classified as osteolytic, osteosclerotic, or mixed types that reflect osteolytic or osteosclerotic changes caused by the highly activated osteoclasts or osteoblasts that occur in bone metastases. In addition, technetium-99m (99mTc) is one of the most important radionuclides in nuclear medicine. 99mTc has frequently been clinically used because (1) it has adequate physical half-life (T1/2 = 6.01 h) for clinical use, (2) the gamma ray energy it emits (141 keV) is appropriate for SPECT imaging, and (3) it can be produced from the radionuclide generator 99Mo/99mTc, which enables generation of 99mTc on demand.

Fig. 1 Chemical structures of bisphosphonates analogs (a) pyrophosphate, (b) MDP, (c) HMDP, (d) EDTMP, and (e) HEDP.
Fig. 1

Chemical structures of bisphosphonates analogs (a) pyrophosphate, (b) MDP, (c) HMDP, (d) EDTMP, and (e) HEDP.

The 99mTc-bisphosphonate complex accumulates in bones because of its high affinity for hydroxyapatite in the bisphosphonate structure. It is assumed that 99mTc-MDP forms a bidentate – bidentate bridge with hydroxyapatite, whereas 99mTc-HMDP must form a bidentate – tridentate bridge because of the presence of an additional hydroxyl group on the central carbon of the C-P-C structure in HMDP and is expected to enhance the hydroxyapatite affinity of the 99mTc complex [6, 7].

The uptake mechanisms of 99mTc-bisphoshonate complexes in bone metastases have not been completely elucidated. One of the factors related to the higher tracer uptake at the metastatic sites is increased vascularity and regional distribution of blood flow associated with the disease. However, regional bone blood flow alone does not explain the increased uptake of 99mTc-bisphoshonate [8]. Other factors are also related to the binding and interaction between the complexes and the bones. It is known that 99mTc-bisphoshonate complexes accumulate at sites of new bone formation or calcification [9, 10]. Kanishi has reported that the accumulation mechanisms might involve both adsorption onto the surface of hydroxyapatite in the bone and incorporation into the crystalline structure of hydroxyapatite [11]. The crystalline structure of hydroxyapatite in newly formed bone is amorphous and has a greater surface area than that in normal bone [12]. Bisphosphonate compounds show significantly higher in vitro adsorption onto amorphous calcium phosphate than onto crystalline forms [8].

99mTc is supplied from the 99Mo/99mTc generator as 99mTcO4. The oxidation state of 99mTc in 99mTcO4 is +7. Bisphosphonate compounds form multiple complexes with reduced 99mTc. By using high-performance liquid chromatography (HPLC), the relative composition of 99mTc-bisphosphonate complexes in a reaction mixture has been found to vary with pH, 99Tc carrier, and oxygen concentrations [13]. Wilson et al. have assumed that 99mTc-bisphosphonate complexes would be a mixture of monomers, oxobridged dimers, and oligomeric clusters with various technetium-oxo core configurations, oxidation states, and ligand coordination numbers [14]. These 99mTc-bisphosphonate complex species have different biodistribution properties in rats. Pinkerton et al. has reported that the smallest, low charge, mononuclear 99mTc-bisphosphonate complex has the greatest uptake in bone lesions and the highest lesion-to-muscle and lesion-to-normal bone ratios in experiments using each isolated complex by HPLC [15]. Although these studies were performed over a quarter century ago, the exact structures and mechanisms of action of 99mTc-bisphosphonate complexes remain unclear.

2.2 Development of novel diagnostic bone-seeking technetium complexes

Although 99mTc-MDP and 99mTc-HMDP are most frequently used as bone scintigraphy agents, their chemical and pharmaceutical properties have not been optimised. As mentioned above, these complexes are not well-defined single-chemical species but rather mixtures of short-chain and long-chain oligomers [13]. In 99mTc-bisphosphonate complexes, the phosphonate groups are used both as ligands for complexation and as carriers of the radionuclide to bone [16], which could decrease the inherent affinity of bisphosphonate for bone. To develop superior bone-seeking radiopharmaceuticals, a more logical design strategy based on conjugation of a stable 99mTc complex with a carrier like bisphosphonate has been proposed by our research group and other groups. I will now discuss some studies that used this drug design, which allowed the ligand and carrier function to operate independently and effectively.

Verbeke et al. have designed and evaluated a 99mTc-L,L-ethylenedicysteine (EC) complex, a renal tracer agent known to have rapid renal excretion, conjugated to bisphosphonate (99mTc-EC-AMDP, Fig. 2a) [17]. 99mTc-EC-AMDP showed a faster blood clearance and a higher bone/blood ratio, which is an index signal/noise (S/N) ratio, relative to those of 99mTc-MDP in animal experiments.

Fig. 2 Chemical structures of 99mTc-complex-conjugated bisphosphonate compounds (a) 99mTc-EC-AMDP, (b) 99mTc-MAG3-HBP, (c) 99mTc-HYNIC-HBP, (d) [99mTc(CO)3(PzNN-BP)], (e) [99mTc(CO)3(PzNN-ALN)], (f) [99mTc(CO)3(PzNN-PAM)], and (g) 99mTc(CO)3-DPA-alendronate.
Fig. 2

Chemical structures of 99mTc-complex-conjugated bisphosphonate compounds (a) 99mTc-EC-AMDP, (b) 99mTc-MAG3-HBP, (c) 99mTc-HYNIC-HBP, (d) [99mTc(CO)3(PzNN-BP)], (e) [99mTc(CO)3(PzNN-ALN)], (f) [99mTc(CO)3(PzNN-PAM)], and (g) 99mTc(CO)3-DPA-alendronate.

Our research group developed stable 99mTc-complex-conjugated bisphosphonate compounds: 99mTc-mercaptoacetylglycylglycylglycine (MAG3)-conjugated bisphosphonate (99mTc-MAG3-HBP, Fig. 2b) and 99mTc-6-hydrazinonicotinic acid (HYNIC), with tricine and 3-acetylpyridine as co-ligands conjugated to bisphosphonate (99mTc-HYNIC-HBP, Fig. 2c) [18]. In hydroxyapatite-binding experiments in vitro, the binding affinities of 99mTc-complex-conjugated bisphosphonate compounds to hydroxyapatite were higher than that of 99mTc-HMDP. In animal experiments, 99mTc-complex-conjugated bisphosphonate compounds showed higher accumulation in bone than did 99mTc-HMDP, which reflected the findings of hydroxyapatite binding experiments in vitro. However, the blood clearance of 99mTc-MAG3-HBP was delayed because its protein-binding rate in blood was high. Thus, the bone/blood ratio of 99mTc-MAG3-HBP was lower than that of 99mTc-HMDP. The blood clearance of 99mTc-HYNIC-HBP was similar to that of 99mTc-HMDP. The bone/blood ratio of 99mTc-HYNIC-HBP was higher than that of 99mTc-HMDP.

Palma et al. developed a 99mTc-tricarbonyl complex, which is anchored by a pyrazolyl (Pz)-containing ligand, conjugated to the bisphosphonate compounds ([99mTc(CO)3(PzNN-BP)], [99mTc(CO)3(PzNN-ALN)], and [99mTc(CO)3(PzNN-PAM)], Figs. 2d–2f) [19, 20]. The ligands N2S2 and N3S have classically been some of the most useful ligands for complexation of 99mTc complexes. The 99mTc complexes have a (TcO)3+ core with technetium in its +5 oxidation state. In contrast, the 99mTc-tricarbonyl complex has a [Tc(CO)3]+ core with technetium in its +1 oxidation state. 99mTc-tricarbonyl complexes, which are compact and kinetically inert, could be formed from the [Tc(CO)3]+ core with suitable ligands [21]. Palma et al. have confirmed the structures of 99mTc-tricarbonyl-complex-conjugated bisphosphonate compounds by using reversed-phase HPLC analyses. The analyses showed that the compounds had retention times identical to those of the corresponding nonradioactive rhenium (Re) complexes, which revealed the structural analogies because nonradioactive technetium does not exist (99Tc is also a radionuclide). In animal experiments [99mTc(CO)3(PzNN-BP)] showed moderate bone uptake, but the uptake was lower than that of 99mTc-MDP. In contrast, the bone accumulations of [99mTc(CO)3(PzNN-ALN)] and [99mTc(CO)3(PzNN-PAM)] were high and comparable to that of 99mTc-MDP. At 4 h after injection of tracers, [99mTc(CO)3(PzNN-ALN)] and [99mTc(CO)3(PzNN-PAM)] showed higher bone/blood and bone/muscle ratios because they had faster clearance than that of 99mTc-MDP. The differences in bone accumulation among the 99mTc-tricarbonyl complex-conjugated bisphosphonate compounds could be derived from the existence of a hydroxyl group at the central carbon of the P-C-P structure in bisphosphonate compounds because some authors have described that bisphosphonate compounds containing the hydroxyl group have higher affinity for bone [2224].

A 99mTc-tricarbonyl complex-conjugated bisphosphonate that has a structure similar to that of [99mTc(CO)3(PzNN-ALN)] but with dipicolylamine (DPA) was developed by de Rosales et al. and used as a ligand for complexation [99mTc(CO)3-DPA-alendronate, Fig. 2g] [25]. 99mTc(CO)3-DPA-alendronate showed higher affinity for hydroxyapatite than did 99mTc-MDP in in vitro experiments. In animal experiments, the bone accumulation of 99mTc(CO)3-DPA-alendronate was as high as that of 99mTc-MDP.

As mentioned above, some 99mTc-complex-conjugated bisphosphonate compounds have shown superior biodistribution of radioactivity as bone imaging agents relative to that of the existing bone scintigraphic agents, 99mTc-complex, 99mTc-MDP, and 99mTc-HMDP. Consequently, the strategy and concept of 99mTc-complex-conjugated bisphosphonates could be promising for the diagnosis of bone disorders, such as bone metastases.

2.3 Development of novel diagnostic bone-seeking gallium complexes for PET

Presently, 68Ga has attracted much attention as a radionuclide for PET because its radiophysical properties are useful clinically, particularly as a 68Ge/68Ga generator-produced radionuclide with a half-life (T1/2 ) of 68 min [26]. Its production does not require an expensive cyclotron, and 68Ga can be produced on demand in a hospital. Indeed, because the half-life of the parent nuclide 68Ge is long (T1/2 = 270.8 d), the lifespan of the 68Ge/68Ga generator also must be long.

The above-mentioned concept of a stable complex-conjugated bisphosphonate could also be applicable to 68Ga complexes. To develop superior PET tracers for diagnosis of bone disorders, some kinds of radiogallium complex-conjugated carriers for delivery to bone have been reported.

We developed a 67Ga-1, 4, 7, 10-tetraazacyclododecane-1, 4, 7, 10-tetraacetic acid (DOTA) complex-conjugated bisphosphonate (67Ga-DOTA-Bn-SCN-HBP, Fig. 3a) because DOTA forms a stable complex with gallium [27]. Although the aim was to develop a superior bone-seeking 68Ga-labeled agent for PET, 67Ga was used in the initial basic study because of its longer half-life (T1/2 = 78 h). In animal experiments, 67Ga-DOTA-Bn-SCN-HBP accumulated rapidly and highly in bone but was rarely observed in tissues other than bone. Consequently, the bone/blood ratio of 67Ga-DOTA-Bn-SCN-HBP was comparable to that of 99mTc-HMDP.

Fig. 3 Chemical structures of 67/68Ga complex-conjugated bisphosphonate compounds (a) 67Ga-DOTA-Bn-SCN-HBP, (b) 68Ga-BPAMD, (c) 68Ga-NOTA-BP, and (d) 67Ga-DOTA-(Asp)n.
Fig. 3

Chemical structures of 67/68Ga complex-conjugated bisphosphonate compounds (a) 67Ga-DOTA-Bn-SCN-HBP, (b) 68Ga-BPAMD, (c) 68Ga-NOTA-BP, and (d) 67Ga-DOTA-(Asp)n.

Fellner et al. have reported a human study of 68Ga-DOTA-conjugated bisphosphonate (68Ga-BPAMD, Fig. 3b) [28]. 68Ga-BPAMD showed high uptake in osteoblastic metastatic lesions in a prostate cancer patient. The maximum standardised uptake values (SUVmax) were 77.1 and 62.1 in the 10th thoracic and L2 vertebra, respectively, for 68Ga-BPAMD; the respective values were 39.1 and 39.2 for 18F-fluoride, which is a typical bone imaging agent for PET. Basic experiments on 68Ga-BPAMD using µ-PET and a bone metastasis rat model have been also reported [29]. 68Ga-BPAMD showed higher accumulation in metastatic bone lesions than in healthy bone of the same animal (contrast factor = 3.97 ± 1.82).

Furthermore, Suzuki et al. have developed 68Ga-1,4,7-triazacyclononane-1,4, 7-triacetic acid (NOTA)-conjugated bisphosphonate (68Ga-NOTA-BP, Fig. 3c) [30]. Triazamacrocyclic ligands may be more suitable for gallium complexation because of their high conformational and size selectivity. Actually, it has been reported that NOTA forms highly stable chelates and allows faster incorporation of gallium at lower temperatures than DOTA [31]. In animal experiments using Wistar rats, 68Ga-NOTA-BP showed faster clearance and a higher bone/blood ratio than did 99mTc-MDP and 18F-fluoride. Moreover, in a PET study using a mouse model of bone metastasis, 68Ga-NOTA-BP highly accumulated in osteolytic lesions in the tibia.

Next, we investigated acidic amino acids as carriers of radionuclides to bone. Several major noncollagenous bone proteins, such as osteopontin and bone sialoprotein, contain repeating sequences of acidic amino acids, such as Asp or Glu, in their structures [3234]. It has been reported that polyglutamic and polyaspartic acids have high affinities for hydroxyapatite and could be used as carriers for drug delivery to bones [3537]. Recently, we reported 67Ga-DOTA-conjugated L-Asp peptides [67Ga-DOTA-(Asp)n, Fig. 3d] with varying peptide lengths (n = 2, 5, 8, 11, or 14) [38]. The binding affinities for hydroxyapatite of 67Ga-DOTA-(Asp)n depended on their peptide lengths; longer peptides had higher affinities for hydroxyapatite. In biodistribution experiments in normal mice, 67Ga-DOTA-(Asp)8, 67Ga-DOTA-(Asp)11, and 67Ga-DOTA-(Asp)14 showed selectively high accumulation in bones (10.5 ± 1.5, 15.1 ± 2.6, and 12.8 ± 1.7% ID/g, respectively). Although the bone accumulation of 67Ga-DOTA-(Asp)n was lower than that of 67Ga-DOTA-Bn-SCN-HBP, which suggested that bisphosphonates have higher affinities for bone than do polyaspartic acids, 67Ga-DOTA-(Asp)n showed faster blood clearance than did 67Ga-DOTA-Bn-SCN-HBP. Accordingly, the bone/blood ratios of 67Ga-DOTA-(Asp)11 and 67Ga-DOTA-(Asp)14 were comparable to that of 67Ga-DOTA-Bn-SCN-HBP.

These results suggest that not only bisphosphonate molecules but also acidic amino acid peptide sequences could be useful as carriers of radionuclides to bones. Moreover, the use of radiometal complex-conjugated carrier molecules for delivery to bones could be a useful approach for the development of 68Ga PET tracers for bone disorders such as bone metastases.

2.4 Bone-seeking radiopharmaceuticals for palliative therapy of bone metastases

Gamma ray emitting radionuclide-and positron emitting radionuclide-labeled bone-seeking agents are used for the diagnosis of bone disorders such as bone metastases. Bones are one of the most common organs affected by metastatic cancer because of the presence of numerous growth factors in them [39, 40]. Especially, certain cancers, such as breast cancer, lung cancer, and prostate cancer, have a tendency to metastaize to bones. Most cancer patients in late phases of bone metastases suffer severe pain, but the pathophysiology is not well understood, and multiple mechanisms have been postulated. Control of metastatic bone pain is very important for improving patients’ quality of life [4143]. As palliative treatments, nonsteroidal anti-inflammatory drugs (NSAIDs) are the first option for most patients, followed by progression to opioids as the intensity of pain rises by using a three-step “ladder” proposed by the WHO for cancer pain relief. However, it is difficult to control pain in patients with bone metastases. After the initial standard palliative treatment, about half of these patients continue to suffer from substantial bone pain [44]. Moreover, although efforts are made to decrease the side effects of drugs used for pain palliation, these drugs can produce unwanted side effects, such as gastrointestinal ulceration, neutropenia, enhanced bleeding, and disruptions in renal function in the case of NSAIDs, and nausea, sedation, and constipation in the case of opioids. Localised radiation therapy from an external source is an effective method for palliation of metastatic bone pain [45]. Localised radiation therapy decreases metastatic bone pain in many patients. However, because most patients with bone metastases have multiple metastatic lesion sites, treating patients, bone metastases with multiple metastatic lesions with localised radiation therapy is not easy. In that case, internal radiotherapy using bone-seeking compounds labeled with high-energy beta or alpha particle-emitter radionuclides has been shown to be an effective alternative that has fewer side effects than those associated with other treatments [46]. Pain palliation is achieved from beta or alpha particles emitted from radionuclides, but the mechanism of this pain palliation has not been elucidated. Because beta or alpha particles cause damage to tumour cells, palliation effects might occur because of a reduction of mechanical pressure. However, the palliative effects are usually observed before the tumour mass is reduced. Because it is known that lymphocytes, which are radiation-sensitive cells, secrete many kinds of cytokines related to pain, palliation effects could be derived from not only reduction of tumour cells but also damage to lymphocytes in the tumour tissue.

I will next discuss the approved radiopharmaceuticals and recent research related to radiolabeled compounds for palliative therapy. 89SrCl2 (Metastron®) was the first radiopharmaceutical approved by the FDA for the palliation of metastatic bone pain. Strontium (Sr) and calcium (Ca) are alkaline earth metals that are members of family IIA on the periodic table. It is known that the characteristics of Sr and Ca are similar and that Sr accumulates at sites of high osteoblastic activity through incorporation into mineralizing collagen during new bone formation [47]. 89Sr has a long physical half-life of 50.5 d and emits high-energy beta particles, with a maximum energy of 1.46 MeV, and 0.01% gamma rays, with an energy of 910 keV. The usual dose of 89SrCl2 is 148 MBq (4 mCi) or 1.5–2.2 MBq/kg body weight. It has been reported that there is no dose-dependence for pain relief [48]. There is a lot of data on the palliative effects of 89SrCl2 for breast and prostate cancer patients with metastatic bone pain. Review articles have reported pain relief rates ranging from 57% to 92% [4951]. Palliation effects are usually observed within 6 weeks after intravenous injection of 89SrCl2, and the mean duration of pain relief is approximately 6 months [52].

Samarium-153 (153Sm) emits beta particles with maximum energies of 0.81 MeV (20%), 0.71 MeV (49%), and 0.64 MeV (30%) and a 28% abundance of gamma rays with an energy of 103 keV and can be used for imaging, unlike 89Sr. 153Sm has a physical half-life of 46.3 h. 153Sm-EDTMP (Quadramet®), which has also been approved by the FDA, is a complex of 153Sm with ethylenediaminetetramethylene phosphonic acid (EDTMP: lexidronam, Fig. 1d), which is a tetraphosphonate chelator with high affinity for bone. Because the biodistribution of 153Sm-EDTMP is similar to that of 99mTc-MDP, the dosimetry of 153Sm-EDTMP could be predicted using 99mTc-MDP bone scintigraphy [53]. The blood clearance of 153Sm-EDTMP is very rapid, and the compound is excreted via the kidney into the urine [54]. The blood clearance is applicable to a biexponential model with an estimated half-life of 5.5 ± 1.1 and 65.4 ± 9.6 min [55]. About 50% of the injected dose is excreted into the urine at 6–7 h after injection. Most of the remaining dose is accumulated in bones. There is little accumulation in soft tissue, such as the liver, and less than 1% of the injected dose remains in the blood. The usual dose of 153Sm-EDTMP is 37 MBq/kg (1 mCi/kg). Review articles have reported pain relief rates ranging from 62% to 84% [4951]. When 89SrCl2– and 153Sm-EDTMP-treated groups of prostate cancer patients with bone metastases were compared, no statistical differences were observed in response rates and side effects [56].

Radium-223 chloride (223RaCl2), in which 223Ra is the alpha particle-emitting radionuclide, was approved by the FDA and European Medicines Agency (EMA) in 2013. Radium (Ra) also is an alkaline earth metal and a member of family IIA in the periodic table, as are Ca and Sr. Among Ra isotopes, 223Ra has a suitable half-life of 11.4 d for therapy and decays through a chain of daughter nuclides with an emitted total energy of approximately 28 MeV, with most of the energy released as alpha particles. In a phase III randomised trial (Alpharadin in Symptomatic Prostate Cancer Patients: ALSYMPCA), surprisingly, 223RaCl2 significantly improved overall survival in castration-resistant prostate cancer patients with bone metastases [57, 58]. Moreover, 223RaCl2 is associated with a low incidence of myelosuppression, which is assumed to be the major side effect and could be the dose-limiting factor. 223RaCl2 is the first alpha particle-emitting radiopharmaceutical approved for clinical use and has been demonstrated to be an effective therapy for bone metastases; therefore, it is currently attracting much attention.

2.5 Development of novel bone-seeking complexes for palliation therapy of bone metastases

Rhenium has chemical properties similar to those of technetium because both elements are members of family VIIA in the periodic table. Of the rhenium isotopes, there are two radionuclides, 186Re and 188Re, that are useful for radionuclide therapy [59]. Both rhenium radionuclides emit not only high-energy beta particles for therapy but also gamma rays for imaging: 186Re (T1/2 = 3.7 d, βmax = 1.07 MeV, γ = 137 keV) and 188Re (T1/2 = 17.0 h, βmax = 2.12 MeV, γ = 155 keV). Furthermore, because 188Re is a daughter nuclide of 188W (T1/2 = 60 d), 188Re is obtained from an in-house alumina-based 188W/188Re generator, similar to a 99Mo/99mTc generator [60].

In the case of the use of rhenium radionuclides for bone-seeking radiophar-maceuticals in a manner similar to that of 99mTc-MDP and 99mTc-HMDP, it has been reported that rhenium forms complexes with some bisphosphonate derivatives as ligands. 186/188Re-1-hydroxyethylidene-1, 1-diphosphonate (186/188Re-HEDP), which is a 186/188Re-complex with HEDP (Fig. 1e), has been evaluated in clinical research [6163]. Although the chemical characteristics of rhenium and technetium are similar, rhenium is more easily oxidised than is technetium [64], and the stability of 186Re-HEDP is lower than that of 99mTc-bisphosphonate complexes [65]. Some studies have shown that gastric accumulation of radioactivity was observed after injection of 186Re-HEDP in patients with bone metastases [66, 67]. It is known that 186/188ReO4accumulates in the stomach, as does 99mTcO4, and it is thought that this occurs because of the in vivo instability of 186Re-HEDP [68]. Moreover, similar to the phosphonate groups in 99mTc-MDP and 99mTc-HMDP, those in 186/188Re-HEDP are used as both a ligand for complexation and a carrier to bone, which may reduce the inherent affinity of HEDP to bones.

To develop superior 186/188Re-labeled bone-seeking radiopharmaceuticals, I assumed that the above-mentioned concept of a stable complex-conjugated bisphosphonate would be more useful. Therefore, our research group designed and evaluated 186Re-monoaminemonoamidedithiol (MAMA)- and 186Re-mercaptoacetylglycylglycylglycine (MAG3)-conjugated bisphosphonate compounds (186Re-MAMA-BP, 186Re-MAMA-HBP, 186Re-MAG3-HBP; Figs. 4a–c) and reported our findings [24, 69–72]. When 186Re-complexes were incubated in buffered solution at 37°C, the Re-complex-conjugated bisphosphonate compounds, 186Re-MAMA-HBP, 186Re-MAMA-BP, and 186Re-MAG3-HBP, were more stable than 186Re-HEDP, as expected. In biodistribution experiments in mice, little gastric accumulation of radioactivity was observed after injection of 186Re-MAMA-HBP, 186Re-MAMA-BP, and 186Re-MAG3-HBP. The drug design of Re-complex-conjugated bisphosphonates led to better stability in vitro and in vivo. Among these 186Re-complex-conjugated bisphosphonate compounds, 186Re-MAG3-HBP showed the most favourable biodistribution characteristics as a bone-seeking agent, such as high and selective bone accumulation. These favourable biodistribution characteristics could be related to high hydrophilicity (log P value:, –2.68 ± 0.01) and the introduction of a hydroxyl group to the central carbon of the bisphosphonate P-C-P structure. The therapeutic potential of 186Re-MAG3-HBP for palliation of metastatic bone pain in a rat model of bone metastasis was evaluated. Compared with untreated control group rats, 186Re-MAG3-HBP-treated rats showed significant palliation effects, as assessed by the hind paw withdrawal response to stimulation with von Frey filaments [73], and the palliation effects of 186Re-MAG3-HBP tended to be higher than those of 186Re-HEDP. Although 186Re-HEDP did not inhibit tumour growth, 186Re-MAG3-HBP significantly inhibited tumour growth.

Because one of the 99mTc-complex-conjugated bisphosphonate compounds, 99mTc(CO)3-DPA-alendronate, has been introduced above, the same ligand was used to design and evaluate 188Re(CO)3-DPA-alendronate by the same group (Fig. 4d) [74]. 188Re(CO)3-DPA-alendronate also showed higher in vitro stability than did 188Re-HEDP, which oxidised to 188ReO4– (up to 75%) after incubation in PBS for 48 h at 37°C. In imaging experiments, 188Re(CO)3-DPA-alendronate showed superior biodistribution of radioactivity than did 188Re-HEDP, i.e. 188Re(CO)3-DPA-alendronate highly accumulated in metabolically active bone, such as joints with low soft-tissue uptake.

Fig. 4 Chemical structures of 186/188Re complex-conjugated bisphosphonate compounds (a) 186Re-MAMA-BP, (b) 186Re-MAMA-HBP, (c) 186Re-MAG3-HBP, and (d) 188Re(CO)3-DPA-alendronate.
Fig. 4

Chemical structures of 186/188Re complex-conjugated bisphosphonate compounds (a) 186Re-MAMA-BP, (b) 186Re-MAMA-HBP, (c) 186Re-MAG3-HBP, and (d) 188Re(CO)3-DPA-alendronate.

These results indicate that the concept of stable 186Re-complex-conjugated bisphosphonates could be very useful and that novel 186Re-complex-conjugated bisphosphonate complexes could be attractive candidates as palliative agents in metastatic bone pain.

3 Radio-complexes for imaging of apoptosis

It is known that apoptosis is associated with maintaining homeostasis, most diseases, and responses to therapy. Therefore, imaging of apoptotic cells could help with elucidation of disease mechanisms and with early detection of therapeutic effects. Next, I will discuss radiocomplexes for imaging of apoptosis, their drug design, preclinical studies, and applications in medicine.

3.1 Technetium-labeled annexin A5

Phosphatidylserine (PS) exists on the intracellular face of the cell membranes of normal cells. When apoptosis occurs, the lipid distribution of the plasma membrane changes so that PS is exposed to the outside of the cell membrane. Therefore, PS could be a target for imaging of apoptosis, and the typical compound of PS-targeted carriers is annexin A5, which is a 36-kDa human protein with a nanomolar affinity for membrane-bound PS [7577]. For imaging of apoptosis, many researchers have developed and evaluated radiolabeled annexin A5 compounds.

3.2 99mTc-4, 5-bis(thioacetamido)pentanoyl-annexin A5 (099mTc-BTAP-annexin A5)

As mentioned above, 99mTc is an ideal radionuclide for clinical use because of its physical properties. However, because most proteins and polypeptides do not possess chelation sites for formation of technetium complexes, a ligand for complexation with technetium should be introduced into proteins and polypeptides. Tetradentate chelators, such as N3S and N2S2 coordination molecules, could form stable square pyramidal 99mTc complexes with a [Tc=O]3+ core in which technetium is in the oxidation state +V. Among the 99mTc-labeled annexin A5 compounds for imaging of apoptosis, 99mTc-4, 5-bis(thioacetamido)pentanoyl (BTAP)-annexin A5 (Fig. 5a), which has a N2S2 ligand for complexation with 99mTc, was reported to be the first 99mTc-labeled annexin A5 compound. 99mTc-BTAP-annexin A5 was used as an imaging agent for transplant rejection of cardiac transplantation by Narula et al. When 99mTc-BTAP-annexin A5 was injected into cardiac allograft recipients, 99mTc-BTAP-annexin A5 accumulated in the hearts of some recipients who showed at least moderate transplant rejection. These results suggested that 99mTc-labeled annexin A5 was useful for noninvasive imaging of apoptosis [78]. However, N2S2 ligands, such as BTAP, should require harsh conditions for preparation of 99mTc complexes because the protecting groups for thiol must be deprotected just before radiolabeling. Thus, 99mTc-BTAP-annexin A5 was prepared by a preformed-chelate approach. The preformed-chelate approach means that the conjugation between the 99mTc-BTAP complex and annexin A5 is performed after the complexation of 99mTc with the BTAP ligand. The preformed-chelate approach is complicated and has low radiochemical yields because multiple steps and purification are needed. Easy 99mTc labeling of annexin A5 is required for routine clinical use.

Fig. 5 Structures of (a) 99mTc-BTAP-annexin A5, (b) 99mTc-HYNIC-annexin A5, and (c) 99mTc-C3(BHam)2-annexin A5.
Fig. 5

Structures of (a) 99mTc-BTAP-annexin A5, (b) 99mTc-HYNIC-annexin A5, and (c) 99mTc-C3(BHam)2-annexin A5.

3.3 99mTc-HYNIC-annexin A5

In 1998, Blankenberg et al. reported 99mTc-HYNIC-annexin A5 (Fig. 5b), for imaging of apoptosis [79]. HYNIC is one of the most familiar ligands for labeling of peptides and proteins with 99mTc. HYNIC forms a mixed ligand complex with 99mTc and the proper coligands. In the complex, it has been reported that HYNIC works as a monodentate or bidentate ligand [80]. Several coligands, such as glucoheptonate, tricine, ethylene diamine diacetic acid (EDDA), and ternary ligand systems containing tricine and water-soluble phosphines or tricine and imine-N-containing heterocycles have been reported. Among these coligands, (tricine)2 has been frequently used for 99mTc labeling of proteins because the 99mTc-(HYNIC)(tricine)2 complex can be prepared under mild conditions with high radiochemical yields in a short reaction time; specifically, 99mTc-(HYNIC) (tricine)2 has been obtained without any purification after a one-step reaction [81, 82]. Presently, 99mTc-HYNIC-annexin A5 with (tricine)2 as coligands is the gold standard among agents used for imaging of apoptosis in nuclear medicine. Because easy labeling has helped researchers with imaging of apoptosis in studies using 99mTc-HYNIC-annexin A5, many of these studies have been conducted in the clinic and as basic research [83]. However, 99mTc-HYNIC-annexin A5 has some disadvantages because of the instability of the 99mTc-(HYNIC)(tricine)2 complex; in particular, 99mTc-HYNIC-annexin A5 shows high uptake and long retention in nontarget tissues, such as the kidney and liver [84].

In basic research, accumulation of 99mTc-HYNIC-annexin A5 in apoptosis-induced Jrukat T-cell lymphoblasts has shown a correlation with the percentage of FITC-labeled annexin A5 labeled cells (r2 = 0.922) [85]. Moreover, many researchers have demonstrated that 99mTc-HYNIC-annexin A5 has high affinity for apoptotic cells and can enable visualisation of apoptosis in various animal models [84, 86–88].

In a clinical study in 2007, 99mTc-HYNIC-annexin A5 imaging before and after initiation of platinum anticancer agent-based chemotherapy in non-small-cell lung cancer patients was reported. All patients who showed increased uptake of 99mTc-HYNIC-annexin A5 in tumours achieved complete or partial responses. The uptake of 99mTc-HYNIC-annexin A5 by tumours correlated with treatment outcome (r2 = 0.86; P = 0.0001) [89].

3.4 99mTc-labeled annexin A5 constructed with histidine residues

In 2002, preparation of mutant annexin A5 with N-terminal extensions constructed with three or six histidine residues for 99mTc labeling was reported [90]. A mutant annexin A5 with six histidine residues in the N-terminal showed higher radiochemical yield, radiochemical purity, and bioactivity than did the mutant annexin A5 with three histidine residues in the N-terminal. This study demonstrated that it was possible to successfully construct annexin A5 with histidine residues in the N-terminal to form specific chelation sites for the 99mTc-carbonyl complex without altering its high affinity.

3.5 99mTc-C3(BHam)2-annexin A5

A novel 99mTc-labeled annexin A5, 99mTc-C3(BHam)2-annexin A5, which has a bis(hydroxamamide) derivative as a bifunctional chelating agent to achieve low uptake and retention in nontarget tissues, was recently reported by my research group (Fig. 5c) [91]. It has been reported that a bis(hydroxamamide) derivative, N, N’-trimethylenedibenzohydroxamide ligand [C3(BHam)2] forms a stable 99mTc complex over a wide pH range under mild reaction conditions within short reaction times [92]. The radiochemical yield is very high even at ligand concentrations as low as 2.5 × 10 6 M. In mice, the radioactivity in liver was not residual after intravenous injection of 99mTc-C3(BHam)2-IgG [93], which indicated that the radiometabolite of 99mTc-C3(BHam)2-IgG could be not residual in metabolic organs. In the case of annexin A5, I assumed that there would be less residual radioactivity in metabolic organs after injection of 99mTc-C3(BHam)2-annexin A5 and therefore, designed 99mTc-C3(BHam)2-annexin A5. The bioactivity of 99mTc-C3(BHam)2-annexin A5 was comparable to that of 99mTc-HYNIC-annexin A5. In mouse biodistribution, the uptake by the kidney was lower for 99mTc-C3(BHam)2-annexin A5 than for 99mTc-HYNIC-annexin A5. Moreover, the radioactivity of metabolic organs, such as the liver and kidney, after injection of 99mTc-C3(BHam)2-annexin A5 gradually decreased, whereas there was residual radioactivity in metabolic organs after the injection of 99mTc-HYNIC-annexin A5. In therapeutic experiments, tumour growth in mice treated with 5-fluorouracil (5-FU) was significantly inhibited. Accumulation of 99mTc-C3(BHam)2-annexin A5 in tumours significantly increased at 24 h after 5-FU treatment. The accumulation of 99mTc-C3(BHam)2-annexin A5 correlated positively with the counts of terminal dUTP nick-end labeling (TUNEL)-positive cells. Moreover, the intratu-moral accumulations of 99mTc-C3(BHam)2-annexin A5 by autoradiography significantly correlated with TUNEL-staining positive cells in corresponding sections (r = 0.716, P < 0.001, Fig. 6).

Fig. 6 Representative (a) autoradiographic images and (b, c) TUNEL-staining images for adjacent tumour sections from mice treated with 5-FU. (d) Correlation between the number of TUNEL-positive cells in each grid (0.45 mm × 0.55 mm) of a tumoural section and 99mTc-C3(BHam)2-annexin A5 accumulation (%dose) determined by autoradiography in each corresponding grid of an adjacent section from mice treated with 5-FU.
Fig. 6

Representative (a) autoradiographic images and (b, c) TUNEL-staining images for adjacent tumour sections from mice treated with 5-FU. (d) Correlation between the number of TUNEL-positive cells in each grid (0.45 mm × 0.55 mm) of a tumoural section and 99mTc-C3(BHam)2-annexin A5 accumulation (%dose) determined by autoradiography in each corresponding grid of an adjacent section from mice treated with 5-FU.

3.6 68Ga-labeled annexin A5

A protein labeling system using 68Ga and a sulfhydryl-derivatised chelator, 2, 2′-(7-(1-carboxy-4-(2-mercaptoethylamino)-4-oxobutyl)-1, 4,7-triazonane-1, 4-diyl)diacetic acid (NODA-GA-T), was reported in 2011 [94]. In that study, 68Ga-labeled annexin A5 was prepared within only 15 min and accumulated in the apoptotic area of myocardial infarctions in a PET study using an animal model. In the same year, site-specific 68Ga-labeled annexin A5 compounds, which are variants of annexin A5 containing a single cysteine residue at a position of 2 or 165 (Cys2-annexin A5 and Cys165-annexin A5, respectively), have been reported [95]. 68Ga-Cys2-annexin A5 and 68Ga-Cys165-annexin A5 were prepared within approximately 55 min with a 25% radiochemical yield (43% if corrected for decay). Both 68Ga-labeled annexin A5 compounds preserved bioactivity and showed high in vitro stability. The uptakes of 68Ga-Cys2-annexin A5 and 68Ga-Cys165-annexin A5 by tumours were not high but were significantly increased by the treatment of cyclophosphamide and radiation therapy in a tumour model.

Radiolabeled annexin A5 seems to require some time before imaging of apoptosis after injection of radiotracers can be performed. Accordingly, although 68Ga is a promising radionuclide for PET imaging, the half-life of 68Ga may be too short if annexin A5 is used as a carrier to apoptotic cells.

3.7 Summary of radiolabeled annexin A5 for imaging of apoptosis

Quantitative imaging agents for apoptosis are useful tools because imaging enables early determination of therapeutic effects on diseases, even before anatomical changes occur at the lesion site. Selection of an appropriate therapy by using imaging data from an individual patient may be possible. Although some radiolabeled-annexin A5 compounds are in use, their biodistributions are not necessarily ideal. I hope that novel apoptosis imaging agents using other carriers to apoptotic cells that enable superior imaging to that currently available will be developed in future.

4 Summary

Radiometal complexes can be used as radiopharmaceuticals because some show ideal biodistribution as diagnostic or therapeutic agents in nuclear medicine. One of the advantages of radiometal complexes is the high radiochemical yield. Radiolabeled compounds with high radiochemical yields do not need purification after radiolabeling. For example, in the case of generator-produced radionuclides, such as 99mTc and 68Ga, radiolabeling and on-demand administration to patients just after eluting radionuclides from generators in the hospital is useful clinically, especially in emergency cases. Two categories of nuclear medicine applications, bone metastases and apoptosis, have been reviewed here, and radiometal complexes have shown great utility in these fields. Development of new radiometal complexes for nuclear medicine is anticipated in the future.

Acknowledgment

This article is also available in: Jastrząb, Tylkowski, New-Generation Bioinorganic Complexes. De Gruyter (2016), isbn 978-3-11-034880-4.

References

[1] van beek E, Lowik C, van der Pluijm G, Papapoulos S. The role of geranylgeranylation in bone resorption and its suppression by bisphosphonates in fetal bone explants in vitro: A clue to the mechanism of action of nitrogen-containing bisphosphonates. J Bone Miner Res 1999;14:722–9.10.1359/jbmr.1999.14.5.722Search in Google Scholar

[2] McClung M, Harris ST, Miller PD, Bauer DC, Davison KS, et al. Bisphosphonate therapy for osteoporosis: benefits, risks, and drug holiday. Am J Med 2013;126:13–20.10.1016/j.amjmed.2012.06.023Search in Google Scholar

[3] Mari C, Catafau A, Carrio I. Bone scintigraphy and metabolic disorders. Q J Nucl Med 1999;43: 259–67.Search in Google Scholar

[4] Domstad PA, Coupal JJ, Kim EE, Blake JS, DeLand FH. 99mTc-hydroxymethane diphosphonate: a new bone imaging agent with a low tin content. Radiology 1980;136:209–11.10.1148/radiology.136.1.6446106Search in Google Scholar

[5] Subramanian G, McAfee JG, Blair RJ, Kallfelz FA, Thomas FD. Technetium-99m-methylene diphosphonate-a superior agent for skeletal imaging: comparison with other technetium complexes. J Nucl Med 1975;16:744–55.Search in Google Scholar

[6] Bevan JA, Tofe AJ, Benedict JJ, Francis MD, Barnett BL. Tc-99m HMDP (hydroxymethylene diphosphonate): a radiopharmaceutical for skeletal and acute myocardial infarct imaging. II. Comparison of Tc-99m hydroxymethylene diphosphonate (HMDP) with other technetium-labeled bone-imaging agents in a canine model. J Nucl Med 1980;21:967–70.Search in Google Scholar

[7] Jurisson SS, Benedict JJ, Elder RC, Whittle R, Deutsch E. Calcium affinity of coordinated diphosphonate ligand. Single-crystal structure of [(en)2Co(O2P(OH)CH2P(OH)O2)]ClO4·H2O. Implication for the chemistry of technetium-99m-diphosphonate skeletal imaging agents. Inorg Ch8 1983.10.1021/ic00151a013Search in Google Scholar

[8] Francis MD, Ferguson DL, Tofe AJ, Bevan JA, Michaels SE. Comparative evaluation of three diphosphonates: in vitro adsorption (C-14 labeled) and in vivo osteogenic uptake (Tc-99m complexed). J Nucl Med 1980;21:1185–9.Search in Google Scholar

[9] Budd RS, Hodgson GS, Hare WS. The relation of radionuclide uptake by bone to the rate of calcium mineralization. II: Patient studies using 99Tcm-MDP. Br J Radiol 1989;62:318–20.10.1259/0007-1285-62-736-318Search in Google Scholar

[10] Budd RS, Hodgson GS, Hare WS. The relation of radionuclide uptake by bone to the rate of calcium mineralization. I: Experimental studies using 45Ca, 32P and 99Tcm-MDP. Br J Radiol 1989;62:314–7.10.1259/0007-1285-62-736-314Search in Google Scholar

[11] Kanishi D. 99mTc-MDP accumulation mechanisms in bone. Oral Surg Oral Med Oral Pathol 1993;75:239–46.10.1016/0030-4220(93)90100-ISearch in Google Scholar

[12] Galasko CS. Mechanism of uptake of bone imaging isotopes by skeletal metastases. Clin Nucl Med 1980;5:565–8.10.1097/00003072-198012000-00014Search in Google Scholar

[13] Tanabe S, Zodda JP, Deutsch E, Heineman WR. Effect of pH on the formation of Tc(NaBH4)-MDP radiopharmaceutical analogues. Int J Appl Radiat Isot 1983;34:1577–84.10.1016/0020-708X(83)90002-9Search in Google Scholar

[14] Wilson GM, Pinkerton TC. Determination of charge and size of technetium diphosphonate complexes by anion-exchange liquid chromatography. Anal Chem 1985;57:246–53.10.1021/ac00279a057Search in Google Scholar

[15] Pinkerton TC, Cheng KT, Shaw SM, Wilson GM. Influence of complex charge and size on the uptake of 99mTc-diphosphonates in osteogenic tissue. Int J Rad Appl Instrum B 1986;13:49–56.10.1016/0883-2897(86)90251-5Search in Google Scholar

[16] Libson K, Deutsch E, Barnett BL. Structural characterization of a technetium-99-diphosphonate complex. Implications for the chemistry of technetium-99m skeletal imaging agents. J Am Chem Soc 1980;102:2476–8.10.1021/ja00527a066Search in Google Scholar

[17] Verbeke K, Rozenski J, Cleynhens B, Vanbilloen H, de Groot T, et al. Development of a conjugate of 99mTc-EC with aminomethylenediphosphonate in the search for a bone tracer with fast clearance from soft tissue. Bioconjug Chem 2002;13:16–22.10.1021/bc0001600Search in Google Scholar PubMed

[18] Ogawa K, Mukai T, Inoue Y, Ono M, Saji H. Development of a novel 99mTc-chelate-conjugated bisphosphonate with high affinity for bone as a bone scintigraphic agent. J Nucl Med 2006;47:2042–7.Search in Google Scholar

[19] Palma E, Correia JD, Oliveira BL, Gano L, Santos IC, et al. 99mTc(CO)3-labeled pamidronate and alendronate for bone imaging. Dalton Trans 2011;40:2787–96.10.1039/c0dt01396jSearch in Google Scholar PubMed

[20] Palma E, Oliveira BL, Correia JD, Gano L, Maria L, et al. A new bisphosphonate-containing 99mTc(I) tricarbonyl complex potentially useful as bone-seeking agent: synthesis and biological evaluation. J Biol Inorg Chem 2007;12:667–79.10.1007/s00775-007-0215-0Search in Google Scholar PubMed

[21] Lipowska M, He H, Malveaux E, Xu X, Marzilli LG, et al. First evaluation of a 99mTc-tricarbonyl complex, 99mTc(CO)3(LAN), as a new renal radiopharmaceutical in humans. J Nucl Med 2006;47:1032–40.Search in Google Scholar

[22] van Beek E, Hoekstra M, van de Ruit M, Lowik C, Papapoulos S. Structural requirements for bisphosphonate actions in vitro. J Bone Miner Res 1994;9:1875–82.10.1002/jbmr.5650091206Search in Google Scholar PubMed

[23] Meyer JL, Nancollas GH. The influence of multidentate organic phosphonates on the crystal growth of hydroxyapatite. Calcif Tissue Res 1973;13:295–303.10.1007/BF02015419Search in Google Scholar PubMed

[24] Ogawa K, Mukai T, Arano Y, Otaka A, Ueda M, et al. Rhenium-186-monoaminemonoamidedithiol-conjugated bisphosphonate derivatives for bone pain palliation. Nucl Med Biol 2006;33:513–20.10.1016/j.nucmedbio.2006.03.006Search in Google Scholar PubMed

[25] de Rosales RTM, Finucane C, Mather SJ, Blower PJ. Bifunctional bisphosphonate complexes for the diagnosis and therapy of bone metastases. Chem Commun 2009:4847–9.10.1039/b908652hSearch in Google Scholar PubMed PubMed Central

[26] Zhernosekov KP, Filosofov DV, Baum RP, Aschoff P, Bihl H, et al. Processing of generator-produced 68Ga for medical application. J Nucl Med 2007;48:1741–8.10.2967/jnumed.107.040378Search in Google Scholar PubMed

[27] Ogawa K, Takai K, Kanbara H, Kiwada T, Kitamura Y, et al. Preparation and evaluation of a radiogallium complex-conjugated bisphosphonate as a bone scintigraphy agent. Nucl Med Biol 2011;38:631–6.10.1016/j.nucmedbio.2010.12.004Search in Google Scholar

[28] Fellner M, Baum RP, Kubicek V, Hermann P, Lukes I, et al. PET/CT imaging of osteoblastic bone metastases with 68Ga-bisphosphonates: first human study. Eur J Nucl Med Mol Imaging 2010;37:834.10.1007/s00259-009-1355-ySearch in Google Scholar

[29] Fellner M, Biesalski B, Bausbacher N, Kubicek V, Hermann P, et al. 68Ga-BPAMD: PET-imaging of bone metastases with a generator based positron emitter. Nucl Med Biol 2012;39:993–9.10.1016/j.nucmedbio.2012.04.007Search in Google Scholar

[30] Suzuki K, Satake M, Suwada J, Oshikiri S, Ashino H, et al. Synthesis and evaluation of a novel 68Ga-chelate-conjugated bisphosphonate as a bone-seeking agent for PET imaging. Nucl Med Biol 2011;38:1011–8.10.1016/j.nucmedbio.2011.02.015Search in Google Scholar

[31] Riss PJ, Kroll C, Nagel V, Rosch F. NODAPA-OH and NODAPA-(NCS)n: synthesis, 68Ga-radiola-belling and in vitro characterisation of novel versatile bifunctional chelators for molecular imaging. Bioorg Med Chem Lett 2008;18:5364–7.10.1016/j.bmcl.2008.09.054Search in Google Scholar

[32] Oldberg A, Franzen A, Heinegard D. The primary structure of a cell-binding bone sialoprotein. J Biol Chem 1988;263:19430–2.10.1016/S0021-9258(19)77651-0Search in Google Scholar

[33] Butler WT. The nature and significance of osteopontin. Connect Tissue Res 1989;23:123–36.10.3109/03008208909002412Search in Google Scholar PubMed

[34] Oldberg A, Franzen A, Heinegard D. Cloning and sequence analysis of rat bone sialoprotein (osteopontin) cDNA reveals an Arg-Gly-Asp cell-binding sequence. Proc Natl Acad Sci U S A 1986;83:8819–23.10.1073/pnas.83.23.8819Search in Google Scholar PubMed PubMed Central

[35] Kasugai S, Fujisawa R, Waki Y, Miyamoto K, Ohya K Selective drug delivery system to bone: small peptide (Asp)6 conjugation. J Bone Miner Res 2000;15:936–43.10.1359/jbmr.2000.15.5.936Search in Google Scholar PubMed

[36] Yokogawa K, Miya K, Sekido T, Higashi Y, Nomura M, et al. Selective delivery of estradiol to bone by aspartic acid oligopeptide and its effects on ovariectomized mice. Endocrinology 2001;142:1228–33.10.1210/endo.142.3.8024Search in Google Scholar PubMed

[37] Wang D, Miller S, Sima M, Kopeckova P, Kopecek J. Synthesis and evaluation of water-soluble polymeric bone-targeted drug delivery systems. Bioconjug Chem 2003;14:853–59.10.1021/bc034090jSearch in Google Scholar

[38] Ogawa K, Ishizaki A, Takai K, Kitamura Y, Kiwada T, et al. Development of novel radiogallium-labeled bone imaging agents using oligo-aspartic acid peptides as carriers. PLoS One 2013;8:e84335.10.1371/journal.pone.0084335Search in Google Scholar

[39] Yoneda T, Sasaki A, Mundy GR. Osteolytic bone metastasis in breast cancer. Breast Cancer Res Treat 1994;32:73–84.10.1007/BF00666208Search in Google Scholar

[40] Coleman RE. Skeletal complications of malignancy. Cancer 1997;80:1588–94.10.1002/(SICI)1097-0142(19971015)80:8+<1588::AID-CNCR9>3.0.CO;2-GSearch in Google Scholar

[41] Portenoy RK, Payne D, Jacobsen P. Breakthrough pain: characteristics and impact in patients with cancer pain. Pain 1999;81:129–34.10.1016/S0304-3959(99)00006-8Search in Google Scholar

[42] Portenoy RK, Lesage P. Management of cancer pain. Lancet 1999;353:1695–700.10.1016/S0140-6736(99)01310-0Search in Google Scholar

[43] Mercadante S. Malignant bone pain: pathophysiology and treatment. Pain 1997;69:1–18.10.1016/S0304-3959(96)03267-8Search in Google Scholar

[44] Bonica JJ. Importance of effective pain control. Acta Anaesthesiol Scand Suppl 1987;85:1–16.10.1111/j.1399-6576.1987.tb02665.xSearch in Google Scholar

[45] Arcangeli G, Giovinazzo G, Saracino B, D’Angelo L, Giannarelli D, et al. Radiation therapy in the management of symptomatic bone metastases: the effect of total dose and histology on pain relief and response duration. Int J Radiat Oncol Biol Phys 1998;42:1119–26.10.1016/S0360-3016(98)00264-8Search in Google Scholar

[46] Lewington VJ. Bone-seeking radionuclides for therapy. J Nucl Med 46 Suppl 2005;1:38S-47S.Search in Google Scholar

[47] Davis J, Cook ND, Pither RJ. Biologic mechanisms of 89SrCl2 incorporation into type I collagen during bone mineralization. J Nucl Med 2000;41:183–8.Search in Google Scholar

[48] Silberstein EB, Williams C. Strontium-89 therapy for the pain of osseous metastases. J Nucl Med 1985;26:345–8.Search in Google Scholar

[49] Paes FM, Serafini AN. Systemic metabolic radiopharmaceutical therapy in the treatment of metastatic bone pain. Semin Nucl Med 2010;40:89–104.10.1053/j.semnuclmed.2009.10.003Search in Google Scholar

[50] Liepe K, Kotzerke J. Internal radiotherapy of painful bone metastases. Methods 2011;55:258–70.10.1016/j.ymeth.2011.07.001Search in Google Scholar

[51] Paes FM, Ernani V, Hosein P, Serafini AN. Radiopharmaceuticals: when and how to use them to treat metastatic bone pain. J Support Oncol 2011;9:197–205.10.1016/j.suponc.2011.06.004Search in Google Scholar

[52] Laing AH, Ackery DM, Bayly RJ, Buchanan RB, Lewington VJ, et al. Strontium-89 chloride for pain palliation in prostatic skeletal malignancy. Br J Radiol 1991;64:816–22.10.1259/0007-1285-64-765-816Search in Google Scholar

[53] Bianchi L, Baroli A, Marzoli L, Verusio C, Chiesa C, et al. Prospective dosimetry with 99mTc-MDP in metabolic radiotherapy of bone metastases with 153Sm-EDTMP. Eur J Nucl Med Mol Imaging 2009;36:122–9.10.1007/s00259-008-0926-7Search in Google Scholar

[54] Singh A, Holmes RA, Farhangi M, Volkert WA, Williams A, et al. Human pharmacokinetics of samarium-153 EDTMP in metastatic cancer. J Nucl Med 1989;30:1814–8.Search in Google Scholar

[55] Bayouth JE, Macey DJ, Kasi LP, Fossella FV. Dosimetry and toxicity of samarium-153-EDTMP administered for bone pain due to skeletal metastases. J Nucl Med 1994;35:63–9.Search in Google Scholar

[56] Dickie GJ, Macfarlane D. Strontium and samarium therapy for bone metastases from prostate carcinoma. Australas Radiol 1999;43:476–9.10.1046/j.1440-1673.1999.00716.xSearch in Google Scholar

[57] Parker C, Nilsson S, Heinrich D, Helle SI, O’Sullivan JM, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369:213–23.10.1056/NEJMoa1213755Search in Google Scholar

[58] Sartor O, Coleman R, Nilsson S, Heinrich D, Helle SI, et al. Effect of radium-223 dichloride on symptomatic skeletal events in patients with castration-resistant prostate cancer and bone metastases: results from a phase 3, double-blind, randomised trial. Lancet Oncol 2014;15:738–46.10.1016/S1470-2045(14)70183-4Search in Google Scholar

[59] Ferro-Flores G, Arteaga de Murphy C. Pharmacokinetics and dosimetry of 188 Re-pharmaceuticals. Adv Drug Deliv Rev 2008;60:1389–401.10.1016/j.addr.2008.04.008Search in Google Scholar PubMed

[60] Knapp FF, Jr., Beets AL, Guhlke S, Zamora PO, Bender H, et al. Availability of rhenium-188 from the alumina-based tungsten-188/rhenium-188 generator for preparation of rhenium-188-labeled radiopharmaceuticals for cancer treatment. Anticancer Res 1997;17:1783–95.Search in Google Scholar

[61] Ogawa K, Washiyama K. Bone target radiotracers for palliative therapy of bone metastases. Curr Med Chem 2012;19:3290–300.10.2174/092986712801215865Search in Google Scholar PubMed

[62] Liepe K, Runge R, Kotzerke J. The benefit of bone-seeking radiopharmaceuticals in the treatment of metastatic bone pain. J Cancer Res Clin Oncol 2005;131:60–6.10.1007/s00432-004-0625-0Search in Google Scholar PubMed

[63] Finlay IG, Mason MD, Shelley M. Radioisotopes for the palliation of metastatic bone cancer: a systematic review. Lancet Oncol 2005;6:392–400.10.1016/S1470-2045(05)70206-0Search in Google Scholar

[64] Deutsch E, Libson K, Vanderheyden JL, Ketring AR, Maxon HR. The chemistry of rhenium and technetium as related to the use of isotopes of these elements in therapeutic and diagnostic nuclear medicine. Nucl Med Biol 1986;13:465–77.10.1016/0883-2897(86)90027-9Search in Google Scholar

[65] Arano Y, Ono M, Wakisaka K, Uezono T, Akizawa H, et al. Synthesis and biodistribution studies of 186Re complex of 1-hydroxyethylidene-1, 1-diphosphonate for treatment of painful osseous metastases. Radioisotopes 1995;44:514–522.10.3769/radioisotopes.44.8_514Search in Google Scholar

[66] De Winter F, Brans B, Van De Wiele C, Dierckx RA. Visualization of the stomach on rhenium-186 HEDP imaging after therapy for metastasized prostate carcinoma. Clin Nucl Med 1999;24: 898–899.10.1097/00003072-199911000-00022Search in Google Scholar

[67] Limouris GS, Shukla SK, Condi-Paphiti A, Gennatas C, Kouvaris I, et al. Palliative therapy using rhenium-186-HEDP in painful breast osseous metastases. Anticancer Res 1997;17:1767–72.Search in Google Scholar

[68] Lin WY, Hsieh JF, Tsai SC, Yen TC, Wang SJ, et al. A comprehensive study on the blockage of thyroid and gastric uptakes of 188Re-perrhenate in endovascular irradiation using liquid-filled balloon to prevent restenosis. Nucl Med Biol 2000;27:83–7.10.1016/S0969-8051(99)00079-7Search in Google Scholar

[69] Ogawa K, Mukai T, Arano Y, Hanaoka H, Hashimoto K, et al. Design of a radiopharmaceutical for the palliation of painful bone metastases: rhenium-186-labeled bisphosphonate derivative. J Labelled Cmp Radiopharm 2004;47:753–61.10.1002/jlcr.864Search in Google Scholar

[70] Ogawa K, Mukai T, Arano Y, Ono M, Hanaoka H, et al. Development of a rhenium-186-labeled MAG3-conjugated bisphosphonate for the palliation of metastatic bone pain based on the concept of bifunctional radiopharmaceuticals. Bioconjug Chem 2005;16:751–7.10.1021/bc040249wSearch in Google Scholar

[71] Ogawa K, Mukai T, Kawai K, Takamura N, Hanaoka H, et al. Usefulness of competitive inhibitors of protein binding for improving the pharmacokinetics of 186Re-MAG3-conjugated bisphos-phonate (186Re-MAG3-HBP), an agent for treatment of painful bone metastases. Eur J Nucl Med Mol Imaging 2009;36:115–21.10.1007/s00259-008-0925-8Search in Google Scholar

[72] Ogawa K, Mukai T, Asano D, Kawashima H, Kinuya S, et al. Therapeutic effects of a 186Re-complex-conjugated bisphosphonate for the palliation of metastatic bone pain in an animal model. J Nucl Med 2007;48:122–7.Search in Google Scholar

[73] Walker K, Medhurst SJ, Kidd BL, Glatt M, Bowes M, et al. Disease modifying and anti-nociceptive effects of the bisphosphonate, zoledronic acid in a model of bone cancer pain. Pain 2002;100:219–29.10.1016/S0304-3959(02)00040-4Search in Google Scholar

[74] Torres Martin de Rosales R, Finucane C, Foster J, Mather SJ, Blower PJ. 188Re(CO)3-dipicolylamine-alendronate: a new bisphosphonate conjugate for the radiotherapy of bone metastases. Bioconjug Chem 2010;21:811–5.10.1021/bc100071kSearch in Google Scholar PubMed

[75] Vriens PW, Blankenberg FG, Stoot JH, Ohtsuki K, Berry GJ, et al. The use of technetium Tc-99m annexin V for in vivo imaging of apoptosis during cardiac allograft rejection. J Thorac Cardiovasc Surg 1998;116:844–53.10.1016/S0022-5223(98)00446-2Search in Google Scholar

[76] Tait JF, Gibson D, Fujikawa K. Phospholipid binding properties of human placental anticoagulant protein-I, a member of the lipocortin family. J Biol Chem 1989;264:7944–9.10.1016/S0021-9258(18)83133-7Search in Google Scholar

[77] Walker JH, Boustead CM, Koster JJ, Bewley M, Waller DA. Annexin V, a calcium-dependent phospholipid-binding protein. Biochem Soc Trans 20:828–33.10.1042/bst0200828Search in Google Scholar PubMed

[78] Narula J, Acio ER, Narula N, Samuels LE, Fyfe B, et al. Annexin-V imaging for noninvasive detection of cardiac allograft rejection. Nat Med 2001;7:1347–52.10.1038/nm1201-1347Search in Google Scholar PubMed

[79] Blankenberg FG, Katsikis PD, Tait JF, Davis RE, Naumovski L, et al. In vivo detection and imaging of phosphatidylserine expression during programmed cell death. Proc Natl Acad Sci U S A 1998;95:6349–54.10.1073/pnas.95.11.6349Search in Google Scholar PubMed PubMed Central

[80] King RC, Surfraz MB, Biagini SC, Blower PJ, Mather SJ. How do HYNIC-conjugated peptides bind technetium? Insights from LC-MS and stability studies. Dalton Trans 2007:4998–5007.10.1039/b705111eSearch in Google Scholar PubMed PubMed Central

[81] D’Alessandria C, di Gialleonardo V, Chianelli M, Mather SJ, de Vries EF, et al. Synthesis and optimization of the labeling procedure of 99mTc-HYNIC-interleukin-2 for in vivo imaging of activated T lymphocytes. Mol Imaging Biol 2010;12:539–46.10.1007/s11307-009-0285-1Search in Google Scholar PubMed PubMed Central

[82] Hnatowich DJ, Mardirossian G, Rusckowski M, Fogarasi M, Virzi F, et al. Directly and indirectly technetium-99m-labeled antibodies–a comparison of in vitro and animal in vivo properties. J Nucl Med 1993;34:109–19.Search in Google Scholar

[83] Lahorte CM, Vanderheyden JL, Steinmetz N, Van de Wiele C, Dierckx RA, et al. Apoptosis-detecting radioligands: current state of the art and future perspectives. Eur J Nucl Med Mol Imaging 2004;31:887–919.10.1007/s00259-004-1555-4Search in Google Scholar PubMed

[84] Ohtsuki K, Akashi K, Aoka Y, Blankenberg FG, Kopiwoda S, et al. Technetium-99m HYNIC-annexin V: a potential radiopharmaceutical for the in-vivo detection of apoptosis. Eur J Nucl Med 1999;26:1251–8.10.1007/s002590050580Search in Google Scholar PubMed

[85] Blankenberg FG, Katsikis PD, Tait JF, Davis RE, Naumovski L, et al. Imaging of apoptosis (programmed cell death) with 99mTc annexin V. J Nucl Med 1999;40:184–91.Search in Google Scholar

[86] Doue T, Ohtsuki K, Ogawa K, Ueda M, Azuma A, et al. Cardioprotective effects of erythropoietin in rats subjected to ischemia-reperfusion injury: assessment of infarct size with 99mTc-annexin V. J Nucl Med 2008;49:1694–700.10.2967/jnumed.107.050260Search in Google Scholar PubMed

[87] Takei T, Kuge Y, Zhao S, Sato M, Strauss HW, et al. Time course of apoptotic tumor response after a single dose of chemotherapy: comparison with 99mTc-annexin V uptake and histologic findings in an experimental model. J Nucl Med 2004;45:2083–7.Search in Google Scholar

[88] Guo MF, Zhao Y, Tian R, Li L, Guo L, et al. In vivo99mTc-HYNIC-annexin V imaging of early tumor apoptosis in mice after single dose irradiation. J Exp Clin Cancer Res 2009;28:136.10.1186/1756-9966-28-136Search in Google Scholar

[89] Kartachova M, van Zandwijk N, Burgers S, van Tinteren H, Verheij M, et al. Prognostic significance of 99mTc Hynic-rh-annexin V scintigraphy during platinum-based chemotherapy in advanced lung cancer. J Clin Oncol 2007;25:2534–9.10.1200/JCO.2006.10.1337Search in Google Scholar

[90] Tait JF, Smith C, Gibson DF. Development of annexin V mutants suitable for labeling with Tc(I)-carbonyl complex. Bioconjug Chem 2002;13:1119–23.10.1021/bc025545sSearch in Google Scholar

[91] Ogawa K, Ohtsuki K, Shibata T, Aoki M, Nakayama M, et al. Development and evaluation of a novel 99mTc-labeled annexin A5 for early detection of response to chemotherapy. PLoS One 2013;8:e81191.10.1371/journal.pone.0081191Search in Google Scholar

[92] Xu LC, Nakayama M, Harada K, Nakayama H, Tomiguchi S, et al. Synthesis and evaluation of hydroxamamide-based tetradentate ligands as a new class of thiol-free chelating molecules for 99mTc radiopharmaceuticals. Nucl Med Biol 1998;25:295–303.10.1016/S0969-8051(97)00208-4Search in Google Scholar

[93] Xu LC, Nakayama M, Harada K, Kuniyasu A, Nakayama H, et al. Bis(hydroxamamide)-based bifunctional chelating agent for 99mTc labeling of polypeptides. Bioconjug Chem 1999;10:9–17.10.1021/bc980024jSearch in Google Scholar PubMed

[94] Wangler C, Wangler B, Lehner S, Elsner A, Todica A, et al. A universally applicable 68Ga-labeling technique for proteins. J Nucl Med 2011;52:586–91.10.2967/jnumed.110.082198Search in Google Scholar PubMed

[95] Bauwens M, De Saint-Hubert M, Devos E, Deckers N, Reutelingsperger C, et al. Site-specific 68Ga-labeled Annexin A5 as a PET imaging agent for apoptosis. Nucl Med Biol 2011;38:381–92.10.1016/j.nucmedbio.2010.09.008Search in Google Scholar PubMed

Published Online: 2016-5-31

© 2016 Walter de Gruyter Berlin/Boston

Downloaded on 23.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/psr-2016-0005/html?lang=en
Scroll to top button