Abstract
Radioimmunoimaging and therapy has been an area of interest for several decades. Steady progress has been made towards clinical translation of radiolabeled monoclonal antibodies for diagnosis and treatment of diseases.
Tremendous advances have been made in imaging technologies such as positron emission tomography (PET). However, these advances have so far eluded routine translation into clinical radioimmunoimaging applications due to the mismatch between the short half-lives of routinely used positron-emitting radionuclides such as 18F versus the pharmacokinetics of most intact monoclonal antibodies of interest. The lack of suitable positron-emitting radionuclides that match the pharmacokinetics of intact antibodies has generated interest in exploring the use of longer-lived positron emitters that are more suitable for radioimmunoimaging and dosimetry applications with intact monoclonal antibodies.
In this review, we examine the opportunities and challenges of radioimmunoimaging with select longer-lived positron-emitting radionuclides such as 124I, 89Zr and 86Y with respect to radionuclide production, ease of radiolabeling intact antibodies, imaging characteristics, radiation dosimetry and clinical translation potential.
Keywords: PET imaging, radioimmunoimaging, monoclonal antibodies, longer-lived positron-emitters
Introduction and Background
Applications of radiation in medicine have been described now for over 100 years. The use of radiation in medicine branches from many scientific discoveries, most notably the discovery of x-rays in 1895 and its use in surgery (1). Since that time frame, over the next 60 years critical advances in nuclear medicine technology and instrumentation have resulted in methodologies and technologies for the visualization of many of the body’s organs, including liver and spleen scanning, brain tumor localization, and studies of the gastrointestinal tract by the injection of radionuclides (2, 3). The greatest potential use of radiation in medicine is its utility to provide diagnostic information of pathological processes before the outset of structural changes in an organ. In these applications, very small amounts of radioactive material most often labeled or conjugated to “smart” targeting agents such as antibodies, peptides, and small molecules are introduced into the body. These “smart” agents specifically target individual cells instead of just the general tissues or organs, and therefore provide more valuable information about actual pathology. One example of such “smart” targeting agents is the monoclonal antibody (mAb). Currently, 21 monoclonal antibodies (all intact) are approved by the U.S Food and Drugs Administration (U.S.F.D.A) for diagnosis and treatment of various illnesses.
Antibodies in Nuclear Medicine
Currently radiolabeled antibodies are clinically used for numerous applications such as oncology and cardiology (4, 5). Use of radiolabeled antibodies for targeting specific organs in animals have been explored almost half a century ago (6, 7). Radiolabeled antibodies have been used in the clinic for therapeutic and diagnostic purposes now for over 40 years (8, 9). In 1978, Goldenberg and colleagues successfully applied the principles of antibody-antigen binding by visualizing carcinoembryonic antigen (CEA) on tumors of patients with a history of cancer of diverse histopathology by injecting 131I labeled goat IgG targeting CEA. However, the large scale application of radiolabeled antibodies in the clinic was hindered due to low production yields and concerns of toxic immune reactions after injecting antibodies of animal origin into humans. The introduction of hybridoma technology for monoclonal antibody (mAb) production as developed by Kohler and colleagues along with the evolution of recombinant DNA technology has addressed many of these problems (10). Chimeric and humanized monoclonal antibodies and even completely human antibodies are now the standard. Numerous clinical studies have since been reported describing the use of radiolabeled intact antibodies for diagnosis of cancer using γ-scintigraphy and single photon emission tomography (SPECT) imaging (4, 11–13).
In spite of great successes in pre-clinical animal models, the promise of radioimmunoimaging by γ-scintigraphy has not fully lived up to expectations, mostly due to differences in biodistribution and pharmacokinetic characteristics between animals and humans, and limitations of γ-scintigraphy in terms of intrinsic spatial resolution (14). Radionuclides that decay with γ-energies lower than 100 keV produce too much scatter, while γ-energies over 250 keV are difficult to collimate and therefore pose a challenge for quantitative γ-scintigraphy. To overcome the disadvantages posed by γ-scintigraphy attempts were made to exploit the superiorities of positron emission tomography (PET) for radioimmunoimaging. A single positron decay results in two 511 keV photons being emitted at 180 degrees. Most PET cameras contain a circular array of detectors with coincidence circuits designed to capture 511 keV photons emitted in opposite direction and therefore offer much better resolution and counting efficiency as compared to conventional γ-scintigraphy and SPECT cameras (15). However, a major limitation challenging PET radioimmunoimaging was that the half-life of most of the routinely used PET radionuclides such as 18F (t1/2 = 1.8 h) and 11C (t1/2 = 0.3 h). The half-lives of routinely used PET radionuclides simply did not match well with the biological half-lives (up to several days) and pharmacokinetic parameters of slowly localizing intact antibodies. To overcome the initial blood pool uptake and slow localization of the intact antibody, antibody fragments (Fab) were labeled with 18F for PET imaging. However, 18F labeled antibody fragments failed to demonstrate high tumor localization as demonstrated by parent intact antibodies (16, 17). 18F labeled antibody fragments were cleared from the tumor relatively more quickly than intact antibodies and then rapidly excreted through the urinary route delivering high radiation doses to the kidneys (16, 17). Numerous attempts have been made to modify intact antibodies to increase tumor uptake and residence time, and concomitantly decrease kidney uptake for successful PET with 18F (18, 19); however, to date results from these strategies have demonstrated low to moderate success. Another drawback of using 18F as a choice of radionuclide for PET radioimmunoimaging is the cumbersome chemistry involved which requires synthesis of 18F labeled intermediate precursors for indirect labeling of antibodies with 18F (17, 20)
As an alternative to the approach of modifying the antibody to match the physical characteristics of 18F, the use of longer-lived positron emitters has been explored for radioimmunoimaging with intact antibodies (21–23). The half-lives of longer-lived positron emitters such as 124I and 89Zr (shown in Table 1) have a much closer match with the biological half-life of most intact antibodies. Antibodies labeled with longer-lived positron emitters can be imaged for 2–5 days after injection when the radioactivity in the blood pool has been cleared and target to background ratio is higher. In this report, we have examined the potentials and challenges of selected longer-lived positron-emitting radionuclides for radioimmunoimaging with intact antibodies. The criteria applied for selecting these longer-lived positron emitters (Table 1) included previously published reports with antibodies or clinical studies, a potentially suitable half-life (10 hr–140 h) versus biological half-life, an imageable positron emission, available or potentially available conjugation and radiolabeling chemistry, and reasonable or acceptable production logistics and radiation toxicity.
Table 1.
Characteristics of selected longer-lived positron-emitting radionuclides for radioimmunoimaging. Some of the data presented in the table were obtained from references (4, 159) and http://www.nndc.bnl.gov/nudat2/ (Nuclear Structure and Decay Data Searchable Database, National Nuclear Data Center, Brookhaven National Laboratory, USA) accessed on 03/07/2008.
Radionuclide | Half-life | Production | Daughter (half-life) | β+max in MeV (β+ yields) | γ-energies in MeV (yields) | Mean range (mm) | Intrinsic spatial resolution loss (mm) | Human studies |
---|---|---|---|---|---|---|---|---|
18F | 1.83 h |
20Ne(d, α)18F 18O(p, n)18F |
18O (stable) | 0.63 (97 %) | 0.14 (41 %) | 0.69 | 0.7 | Yes |
124I | 100.2 h |
124Te(p, n)124I 124Te(d, 2n)124I 125Te(p, 2n)124I |
124Te (stable) | 2.14 (24 %) | 0.60 (61 %) | 3.25 | 2.3 | Yes |
76Br | 16.0 h |
76Se(p, n)76Br 75As(3He, 2n)76Br |
76Se (stable) | 3.98 (56 %) | 0.55 (74 %) | 5.07 | 5.3 | Yes |
52Mn | 134.2 h |
52Cr(p, n)52Mn 51V(3He, 2n)52Mn 52Cr(3He,t)52Mn |
52Cr (stable) | 0.58 (29 %) | 0.74 (90 %) | 5.00 | 0.6 | Yes |
55Co | 17.5 h |
54Fe(d, n)55Co 56Fe(p, 2n)55Co |
55Fe ( 2.6 y) | 1.50 (76 %) | 0.48 (20 %) | 5.74 | 1.6 | Yes |
66Ga | 9.4 h |
66Zn(p, n)66Ga 63Cu(4He,n)66Ga |
66Zn (stable) | 4.15 (56 %) | 1.03 (36.9 %) | 8.06 | 5.8 | Yes |
72As | 25.9 h | 72Ge(p, n)72As | 72Ge (stable) | 3.32 (88 %) | 0.83 (80 %) | 5.01 | 3.6 | Yes |
64Cu | 12.7 h |
64Ni(p, n)64Cu 68Zn(p, n)64Cu 64Ni(d,2 n)64Cu |
64Ni,64Zn (stable) | 0.66 (18 %) | - | 0.70 | 0.7 | Yes |
86Y | 14.7 h |
86Sr(p, n)86Y natRb(3He, 2n)86Y |
86Sr (stable) | 3.15 (34 %) | 1.08 (83 %) | 2.46 | 1.8 | Yes |
89Zr | 78.4 h |
89Y(p, n)89Zr 89Y(d, 2n)89Zr |
89mY (16 s) | 0.90 (23 %) | - | 1.18 | 1.0 | Yes |
Selected longer-lived positron-emitting radionuclides for radioimmunoimaging
Numerous longer-lived positron-emitting radionuclides can be produced by cyclotrons, accelerators, and reactors. Selected longer-lived positron emitters ranging from halogens to metals are described below with respect to their production, feasibility of use in radiolabeling an intact antibody, biological applications and imaging characteristics.
Iodine-124
Iodine (atomic radius of 1.3 Å and electronegativity of 2.66) belongs to Group VII of the periodic table. 124I is a positron-emitting radionuclide with a complex decay scheme associated with numerous high energy γ-emissions (0.6 MeV, 61 % abundance) and high-energy positron emissions (2.14 MeV, 24 % abundance) as shown in Table 1. 124I (t1/2= 100.2 h) decays to a stable 124Te daughter. The relatively longer half-life makes 124I conducive for labeling intact antibodies for PET imaging over several days after administration. Additionally, the vast experience of radiolabeling proteins with the other Iodine radionuclides, i.e. 131I, 125I, and 123I, promotes a significant level of confidence since existing chemistry and protocols are directly applicable for use with 124I.
Production of 124I
124I can be produced using a conventional medical cyclotron using the 124Te(p,n)124I and 124Te(d,2n)124I reactions [Table 2]. Typically, highly enriched tellurium dioxide is used as the target and 124I is isolated using dry distillation with minimal impurities (24–26). The yield (<0.1mCi/μAh) from the 124Te(p,n)124I reaction is low at 15 MeV (27). Alternatively, 124I can be produced through a 125Te(p,2n)124I reaction with about four times higher yields than the commonly used 124Te(p,n)124I and 124Te(d,2n)124I reactions (24, 27). However, the production of 124I through the 125Te(p,2n)124I reaction requires a medium-high energy cyclotron and impurities are relatively higher than the 124Te(p,n)124I reaction.
Table 2.
Commonly used production routes of selected longed-lived positron-emitting radioimmunoimaging and corresponding yields and radionuclidic impurities (adapted from reference (119)).
Radionuclide | Commonly used production route | Energy range (MeV) | Calculated yield (MBq/μAh) | Radionuclidic impurity |
---|---|---|---|---|
124I |
124Te(p, n)124I 124Te(d, 2n)124I |
12 → 8 14 → 10 |
16 17.5 |
125I (0.1 %) 125I (1.7 %) |
76Br |
76Se(p, n)76Br 75As(3He, 2n)76Br |
15 → 8 18 → 10 |
360 11 |
- 77Br (1.6 %) |
52Mn |
52Cr(p, n)52Mn 52Cr(3He, t)52Mn |
20 → 10 36→ 10 |
0.4 5.6 |
54Mn (0.5 %) 54Mn (0.8 %) |
55Co |
54Fe(d, n)55Co 58Ni(p, n)55Co |
10 → 5 15 → 7 |
32 14 |
56Co, 57Co (0.1 %) 57Co (0.5 %) |
66Ga | 66Zn(p, n)66Ga | 13 → 8 | 433 | - |
72As | 72Ge(p, n)72As | 18 → 8 | 93 | 71As (10 %) |
64Cu |
64Ni(p,n)64Cu 68Zn(p, n)64Cu |
12 → 9 35 → 20 |
240 100 |
61Cu (0.4 %) 67Cu (25 %) |
86Y | 86Sr(p, n)86Y | 14 → 10 | 400 | 87mY (3 %) |
89Zr | 89Y(p, n)89Zr | 12 → 6 | 43 | 88Zr (0.1 %) |
Radiolabeling of antibodies with 124I
Radio-iodinated antibodies have been used for in vivo applications for more than 60 years (6, 7, 28, 29). In the early 1960’s, Hunter and co-workers described a method (Chloramine-T method) to prepare high specific-activity radio-iodinated antibodies using p-toluene sulfonochloramide [Figure 1(a)] (30, 31). Briefly, the Chloramine-T method is an oxidative method which involves exposure of the substrate to Chloramine-T in the presence of NaI for a short time, producing high specific activity proteins labeled with carrier-free radioiodine. However, the major disadvantage of Choramine-T method is the risk of oxidation of thiol groups and protein denaturation due to the presence of high concentrations of strong oxidizing agent which can compromise the intended biological use of the antibody. As an alternative, an enzymatic method using lactoperoxidase as a catalyst was developed for iodination of antibodies (32, 33). Lactoperoxidase catalyzes the oxidation of iodide using hydrogen peroxide as the enzyme substrate and is a milder oxidative agent than Chloramine-T. Due to the continuing concerns of protein denaturation and loss of biological activity by oxidizing agents, a newer technique (Bolton-Hunter method [Figure 1(b)]) was later developed using iodinated 3-(4-hydroxyphenyl)propionic acid N-hydroxysuccinimide ester, which reacts with free amino groups in the protein molecule to attach the radioiodine-labeled groups by amide bonds (34, 35). The Bolton-Hunter method is a non-oxidative technique which is less harsh on the proteins than alternative methods. Similarly, to avoid the interaction of oxidizing agent with the protein, an alternative solid-phase oxidation using 1,3,4,6-tetrachloro-3 alpha,6 alpha-biphenyl glycoluril [Figure 1(c)] was developed and is commonly called as the Iodogen method (36). All of the above methods suffered from the drawback of in vivo deiodination in presence of enzymes. To overcome in vivo deiodination, another method was developed using N-succinimidyl 4-iodobenzoate (PIB) [Figure 1(d)] to specifically label antibodies and proteins (37). This method produced a more stable radio-iodinated antibody as compared to methods like Bolton-Hunter as PIB does not contain an o-phenol moiety which protects PIB derivatives from the deiodinases that can act to delabel o-iodophenols that are formed with the other radio-iodination methods. As a result of increased in vivo stability, the uptake in the thyroid was dramatically reduced when a comparison was made between the same antibody radioiodinated using the PIB and Chloramine-T methods (37). Many monoclonal antibodies are internalized via either clathrin dependent or independent pathways. Antibodies rapidly internalized (within 2–4 hours) via the clathrin-dependent endocytosis pathway are catabolized within lysosomes. Iodotyrosine is known to rapidly exit from the lysosome and the cell after catabolism and as result target to background ratios are poor (38–40). In order to overcome the issue of catabolism of conventional radio-iodinated antibodies, dilactitol-tyramine (DLT) and radioiodinated diethylenetriaminepentaacetic acid-appended peptides have successfully been used to residualize the radio-iodinated antibody within the cells (41–44). As a result of decreased catabolism, the tumor uptake of cell surface binding radio-iodinated antibodies was significantly higher than antibodies radiolabeled with Chloramine-T method (41, 42).
Figure 1.
General scheme and reagents of radio-halogenation of intact antibodies using commonly used methods (a) Chloramine-T method, (b) Bolton-hunter method, (c) Iodogen method and (d) N-succinimidyl 4-iodobenzoate based method.
Detailed methodologies for radio-iodination of proteins can be found in a review article published in Bioconjugate Chemistry (45). Currently, most 124I labeled antibodies are prepared using commercial iodination kits based on some of the above described techniques.
Biological studies with 124I labeled antibodies
In the early 1990’s, murine monoclonal antibody H17E2 recognizing placental alkaline phosphatase (PLAP) was radiolabeled with 124I using the Iodogen method for targeting PLAP on HEp2 human tumor xenografts (21). The 124I labeled H17E2 localized in the tumor for at least 7 days demonstrating the feasibility of using monoclonal antibodies labeled with 124I for tumor localization studies (21). The utility of antibodies labeled with 124I for PET imaging, imaging feasibility, and quantification studies were studied by Pentlow and co-workers (22). In this study, when compared to 18F, the spatial resolution was only slightly degraded while the linearity was the same (22). This technique was translated to in vivo application by measurements of human neuroblastoma tumors in rats which had been injected with 124I labeled 3F8 antibody demonstrating that quantitative PET imaging of 124I labeled antibodies was possible in biological systems. Use of 124I in PET radioimmunoimaging was further demonstrated by c-erb B2 quantification and visualization in tumor xenografts for up to 160 hours using 124I labeled rat monoclonal antibody (ICR12) recognizing the external domain of the human c-erb B2 proto-oncogene product (23). With advances in imaging instrumentation and technology, whole-animal PET studies were performed for non-invasive measurements of tumor vascular endothelial growth factor (VEGF) in animal model using 124I-SHPP-VG76e (46). Similarly, 124I labeled engineered antibodies have been evaluated for in vivo imaging (47, 48).
Numerous applications of 124I in humans for PET imaging have since been reported (49–51). PET with anti-VEGF 124I-HuMV833 was conducted on twenty patients with progressive solid tumors with moderate success (52). A recently published clinical study successfully used a carbonic anhydrase-IX targeted 124I-cG250 antibody to accurately identify clear-cell renal carcinoma and to predict the aggressiveness of the disease, information that could be valuable in treatment decision making (53).
Challenges
124I is available from numerous academic centers and is also available commercially through sources such as IBA Molecular, but tends to be expensive. Current concerns remain high radiation dose and degraded image quality from high-energy γ-emissions, loss of spatial resolution as a result of high-energy positron [Table 1]. The image quality is further degraded due to decrease in tumor/background ratio as a result of intracellular catabolism of 124I. Typically radioiodide localizes in the thyroid delivering high radiation doses and therefore potassium iodide solution has to be infused before protein labeled with 124I infusion to block thyroid uptake.
Bromine-76
Bromine (atomic radius of 1.1 Å and electronegativity of 2.96) also belongs to the Group VII of the periodic table. 76Br has a fairly high abundance (56%) of high-energy β+ emission (3.9 MeV) and decays to a stable 76Se daughter as shown in Table 1. The half-life of 16.2 h also makes 76Br one of the most attractive radionuclides for PET imaging using intact antibodies that exhibit slow clearance. The positron abundance of 76Br is almost double that of 124I [Table 1] and the C-Br bond is stronger than the C-I bond in organic compounds therefore making it less susceptible to in vivo dehalogenation, thereby potentially minimizing concerns regarding background activity and associated toxicity (54). Due to the smaller atomic radius and greater electronegativity, 76Br has greater polarity than 124I. The increased polarity of 76Br can alter biological properties and pharmacokinetics of the biomolecule which can either be unfavorable or favorable depending upon the objective and purpose of the study (55, 56).
Production of 76Br
76Br can be prepared using a low-intermediate energy medical cyclotron. Typically, 76Br is produced through the 76Se(p,n)76Br reaction in which a highly enriched 76Se target is bombarded with 15–16 MeV protons (57). Alternatively, 76Br can also be prepared through the 75As(3He,2n)76Br and natBr(p, xn)76Kr→76Br reactions using high energy (over 50 MeV) protons (58, 59). Purification and recovery of 76Br is performed by dry distillation methods. 76Br produced through alternative routes contains other radionuclide impurities such as 77Br and 75Br which may result in lower yields due to separation and purification. The calculated yield for 76Br produced through the 76Se(p,n)76Br reaction is almost 32 times greater than 76Br produced through the 75As(3He,2n)76Br reaction [Table 2]. Another disadvantage of 76Br produced through the 75As(3He,2n)76Br reaction is the 77Br ( t1/2=57.03 h) impurity.
Radiolabeling of antibodies with 76Br
Numerous antibodies have been brominated using techniques and approaches similar to iodination such as Chloramine-T, enzymatic, N-succinimidyl para-(tri-methylstannyl)benzoate (60–62). Bromination of antibodies by oxidative methods can be more difficult than iodination due to the lower oxidation potential of bromide. Therefore, more powerful oxidizing agents are used for sufficient yield of bromination, potentially proving deleterious to proteins. Recently, a method was described for indirect radiobromination of antibodies to prevent protein denaturation in presence of strong oxidizing agents (63). In this method, indirect labeling was performed using the p-isothiocyanatobenzene derivative of the [76Br]undecahydro-bromo-7,8-dicarba-nido-undecaborate(1−) (76Br-NBI) as a precursor molecule. 76Br-NBI was prepared using the Chloramine-T method and thereafter the coupling was performed without intermediate purification in an “one-pot” reaction (63). This technique resulted in improved intracellular retention of the radiolabeled antibody and decreased the loss of biological activity caused due to direct bromination with oxidizing agents. Similar indirect radio-bromination efforts have been reported using 4-hydroxyphenylethylmaleimide for site-specific bromination of a cysteine containing variant of the anti-HER2 affibody (64).
Biological studies with 76Br labeled antibodies
Proteins have been brominated and evaluated in animals now for over 30 years (60). In the past decade, anti-CEA MAb 38S1 was radio-brominated and evaluated in animals (65, 66). A comparative study using 76Br-38S1, 18FDG and 11C-Met demonstrated the superiority of 76Br-38S1 in successful identification and imaging of liver metastases in nude rats bearing human colon carcinoma (67). In recent years, more studies have been reported with 76Br labeled intact antibodies and engineered antibodies for the purpose of PET imaging (56, 63, 64, 68). In a recent study, an ED-B fibronectin-binding human antibody derivative (L19-SIP) was labeled with 76Br via an enzymatic approach using bromoperoxidase resulting in both radiolabeling yields and immunoreactivity of over 80% (56). This radio-brominated immunoconjugate (standardized uptake value of over 2.4) enabled detailed small-animal PET imaging of tumor neovasculature in F9 tumor-bearing animals (56). However, long retention of radioactivity in blood and very slow renal excretion were also observed, and as a result, the background activity in non-target organs was higher than that reported for the 125I-labeled L19-SIP, illustrating the differences between iodinated and brominated antibody (56).
Clinically, small molecules labeled with 76Br have been successfully used for in vivo quantitative PET neuroimaging (69, 70); however, few if any radio-brominated antibodies have been successfully used in the clinic.
Challenges
Besides the loss of intrinsic spatial resolution of about 5.3 mm due to high-energy positron (3.98 MeV) [Table 1], one of the major problems with 76Br is the availability and large scale production. Enriched 76Se is relatively expensive due to low natural abundance (1 mg/~US $6) although target material recovery and recycling could obviate that aspect. This may be one of the contributing factors hindering commercialization of large quantities of 76Br, although smaller quantities of 76Br are available on request through MDS Nordion and Isotrace Technology. The β+ energy of 76Br is relatively higher than conventional PET radionuclides which may pose concerns of radiation doses for diagnostic purposes; however the same could be viewed as a potential advantage if actually used for therapeutic applications.
Manganese-52
Manganese (atomic radius of 1.8 Å and electronegativity of 1.55) belongs to Group VIIB of the periodic table. 52Mn (t1/2=134.2 h) decays to a stable 52Cr daughter. The half-life and suitable imaging characteristics as a result of optimum positron energy of 0.58 MeV [Table 1] makes 52Mn one of the most attractive, under-utilized longer-lived positron emitters for PET imaging of intact antibodies.
Production
52Mn can be produced using the 52Cr(p,n)52Mn reaction by irradiating a high purity Cr target with 16–17 MeV protons or by 51V(3He,2n)52Mn using a high-energy cyclotron (71–73).
Radiolabeling of antibodies with 52Mn
Although 52Mn seems to be a suitable candidate of radioimmunoimaging, relatively little effort has been made with it to label antibodies. Chelating agents such as dipyridoxyl diphosphate (74), macrocyclic Schiff-base ligands (73), cyclopentadienide (74) and porphyrin derivatives (75) have been suggested for Mn in different oxidation states. However, generation of stable 52Mn(II) labeled radioimmunoconjugates in aqueous conditions remains unclear due to complex redox chemistry.
To date, no peer-reviewed reports have been published describing the production or use of 52Mn labeled antibodies.
Biological studies with 52Mn
Radio-manganese has mostly been used as a cationic perfusion tracer and as a tracer to study the involvement of manganese in degenerative neurological diseases (71, 72, 75).
Challenges
52Mn is one of the positron-emitters that have been understudied in nuclear medicine. Therefore, production, chelation and conjugation chemistry, and biological effects of 52Mn all need to be extensively studied prior to embarking on radioimmunoimaging application studies.
Cobalt-55
Cobalt belongs to the Group VIIIB of the periodic table. Cobalt has an atomic radius of 1.7 Å and electronegativity of 1.88. 55Co decays to a longer-lived 55Fe daughter (t1/2 = 2.6 y) mainly by positron emission of 1.5 MeV (76% abundant) with a half-life of 17.5 h [Table 1].
Production of 55Co
The 54Fe(d,n)55Co reaction has been used to obtain 55Co in good yields (32 MBq/μAh) and acceptable purity of over 99% using a medical cyclotron [Table 2] (76). Alternatively, 55Co can produced through 58Ni(p,n)55Co on medical cyclotron but yields are less than half that of 54Fe(d,n)55Co reaction [Table 2]. No-carrier-added 55Co can be prepared using the 56Fe(p,2n)55Co reaction by bombardment with 28 MeV protons using a cyclotron or 40 MeV protons using a linear accelerator (77, 78). Better yields and decreased impurities of 56Co are achieved when the reaction is carried out using 28 MeV protons and high-purity target material. Typically, radiochemical separation is performed by solvent extraction techniques (78, 79).
Radiolabeling of antibodies with 55Co
In studies performed at Brookhaven National Laboratory, an anti-CEA antibody fragment was labeled with 55Co using cyclohexyl EDTA monoanhydride (CDTA-MA), 4-isothiocyanato-trans-1,2,-diaminocyclohexane N,N,N′,N′-tetraacetic acid (4-ICE) and diethylenetriamine pentaacetic acid dianhydride (DTPA-DA) as bifunctional chelating agents (77). The radiolabeling was carried out at pH 5.5 at room temperature with an incubation time ranging from 2–4 hours. All three of the immunoconjugates were obtained in high labeling yields and demonstrated good serum stability after 4 days (77).
Biological studies with 55Co labeled antibodies
55Co labeled anti-CEA F(ab′)2 using DTPA-DA and 4-ICE as bifunctional chelating agents was evaluated in athymic mice bearing human colon carcinoma LS174T xenografts as potential PET imaging agent (77). In the same study, to evaluate the feasibility of the use of 55Co as a PET tracer, a Jaszczak phantom study was performed on a whole body PET scanner, providing images of satisfactory resolution (77). In vivo biodistribution studies revealed the superiority of 4-ICE as a better 55Co chelating agent as compared to DTPA-DA as the tumor uptake of 4-ICE conjugated antibody was 2.8 times with greater than that of DTPA-DA conjugated antibody.
In the past, 55Co labeled bleomycin has been evaluated for imaging of tumors in pre-clinical and clinical settings (76, 78, 80). Other applications of 55Co include evaluation of renal function (79). However, most applications of 55Co in clinic are in the field of neuro-imaging to assess neuronal damage in stroke and traumatic brain injury (81, 82).
Challenges
Since the initial reports of successful use of 55Co in radioimmunoimaging in early 1990’s, no peer-reviewed reports appear to have been published in over 15 years demonstrating the use of 55Co in radioimmunoimaging. Issues surrounding appropriate chelation chemistry should be addressable extrapolating from the prior reports and use of 1,4,7,10-tetraaminocyclododecane-N,N,N′,N′-tetraacetic acid (DOTA) derivatives might also prove useful in this application. The dissociation kinetics of Co(II)-DOTA complexes have been suggested to be very slow at neutral and slightly acidic pH and therefore may offer better in vivo stability than DTPA complexes (83, 84). Cobalt has been used to determine the mean number of DOTA chelating groups conjugated per antibody using a metal-binding assay (85, 86). Size considerations might also direct one to investigate NOTA. This is an area of research that remains unsettled. Another potential challenge might be the radiation dose from the 56Co impurity and the generated presence of the longer-lived 55Fe daughter (t1/2=2.6 y) (87). It is known that uncomplexed cobalt localizes in the bone and liver which could pose issues related to radiation toxicity in case the 55Co complex is unstable in vivo (88).
Gallium-66
Gallium belongs to Group III of the periodic table. Gallium has an atomic radius of 1.8 Å and electronegativity of 1.81. 66Ga has one of the highest energies of the candidates considered here for PET imaging (Emax=4.15 MeV, 56% abundant). Along with a complex spectrum of γ-rays, 66Ga is one of the more difficult positron-emitters for quantitative PET imaging. However, due its favorable half-life of 9.4 h, although at the shorter end of this criterion, use of 66Ga has been explored for radioimmunoimaging in the past decade (89, 90).
Production of 66Ga
66Ga can be produced using a medical cyclotron employing the 66Zn(p,n)66Ga reaction [Table 2] (91). Alternatively, 66Ga can be produced by 4He bombardment of natural copper targets by means of the 63Cu (4He, n) 66Ga reaction (89, 92, 93). However, this method also yields an impurity in the form of 67Ga (t1/2= 3.26 d). Chemical purification of 66Ga from the copper target using different techniques is described in detail in these published reports (89, 92, 93).
Radiolabeling of antibodies with 66Ga
The co-ordination chemistry of Ga(III) is well established as it is known that Ga(III) readily binds to the harder oxygen donor chelating agents (94). Initially, GaCl3 is complexed as an acetate or a citrate and this weak coordination complex is then used to prepare complexes of higher stability (94).
Monoclonal antimyosin antibody, Imciromab was labeled with 66Ga in high yield (99%) by transcomplexation from an acetate buffer to the DTPA-conjugated protein (89, 90). One of the major concerns of using 66Ga for labeling antibodies is the formation of the insoluble hydrolyzed Ga(OH)3 species above pH 4.5. The radiolabeling had to be carried out in a pH range between 3–4.5 conditions which may potentially degrade intact antibodies. Suitable chelation chemistry for Ga(III) radionuclides that has been evaluated includes EDTA, DTPA derivatives, and DOTA, the latter of which may be adequately stable in vivo within the context of the half-lives of the positron emitting gallium radionuclides (95). A critical parameter for in vivo stability of the Ga(III)-complex is it’s relatively stability to the Ga(III)-transferrin complex. The reported formation constant for Ga(III)-transferrin complex is log K1 = 20.3 (96) and the high plasma concentration of transferrin increases the risks of transchelation. The majority of the Ga(III) complexes used in nuclear medicine are thermodynamically stable and kinetically inert to exchange with transferrin in vivo. However, the complex formed with the chelator 1,4,7-triazacyclononane-N,N′,N″-triacetic acid (NOTA) is exceptionally stable and quite probably will emerge as the agent of choice for use with Ga(III) radionuclides (97, 98). Moreover, NOTA is a more suitable chelating agent for Ga(III) labeled antibodies as the Ga(III)-NOTA complex is formed at room temperature and therefore the protein is exposed to less harsh conditions as compared to heating as in the use of DOTA (99).
Biological studies with 66Ga labeled antibodies
66Ga-DTPA-antimyosin antibody was evaluated in dogs for imaging of myocardial necrosis by PET (89). Most biological applications of 66Ga in molecular imaging have been with either peptides or small molecules as the radiolabeling in acidic conditions is more favorable as compared to use with antibodies (100, 101). The use of 66Ga in humans dates back to 1954 (102), and in that study 66Ga was used to investigate bone disease. As compared to 66Ga, short-lived positron emitter 68Ga (t1/2= 1.1 h) obtained from a 68Ge/68Ga generator has gained immense popularity in the recent years due to availability of commercial 68Ge/68Ga generators and clinical successes with 68Ga labeled somatostatin peptide (103, 104). In spite of such a short half-life, excellent results have been obtained from a 68Ga labeled trastuzumab fragment in studying the tumor response to the heat shock protein 90 (Hsp90) inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) (105, 106). In these studies, a 68Ga labeled Herceptin fragment was successfully used to image the pharmacodynamics of HER2 degradation in response to 17-AAG and was an earlier predictor of tumor response to 17-AAG therapy than 18F-FDG PET (105, 106). However, 68Ga may not be a suitable candidate for imaging with intact antibodies due to their slower pharmacokinetics and tumor localization.
Challenges
Apart from unfavorable antibody radiolabeling procedures, the utility of 66Ga in quantitative radioimmunoimaging is severely impaired by high positron energy (4.15 MeV) which results in loss of intrinsic spatial resolution of about 5.8 mm and additional high-energy γ-emission (1.03 MeV) resulting to poor signal to noise ratio [Table 1]. The half-life of 9.4 h may be relatively longer than existing radionuclides such 18F (t1/2= 1.83 h), but the half-life is shorter with respect to the blood clearance and tumor localization of an intact antibody. Traditionally, gallium (67Ga) is used for bone scans and therefore; 66Ga lost from the chelate (if any) will localize in the bones delivering high radiation dose.
Arsenic-72
Arsenic belongs to Group III of the periodic table and has an atomic radius of 1.3 Å, and electronegativity of 2.18. 72As (t1/2 = 25.9 h) decays to a stable 72Ge daughter with 88% high-energy positron emission [Table 1]. The longer half-life and positron abundance makes 72As one of the more attractive potential candidates of PET radioimmunoimaging. 72As can also serve as a potential imaging and dosimetry surrogate for the β−-emitting 77As (t1/2 = 38.4 h) which can be used for radioimmunotherapy.
Production of 72As
72As can produced by irradiating an enriched germanium target using the 72Ge(p,n)72As reaction, but the radionuclidic purity is low due to the formation of 71As (10%) as shown in Table 2 (107). The target material can be recovered by dry distillation for reprocessing. Alternatively, 72As can be produced from 72Se/72As radionuclide generators based on solid phase extraction and distillation in which 72As can be eluted in aqueous solvents with over 40–70 % yield with less than 0.1 % contamination of 72Se (108–110). However, for generator based production of 72As, the production of 72Se is tedious as it requires over 35 MeV bombardment of natural germanium target with helium ions. Another limitation of the 72Se/72As radionuclide generator is the relatively short half-life of 72Se (8.40 d).
Radiolabeling of antibodies by 72As
Radio-arsenic (74As and 77As) has been used to label the phosphatidylserine targeting chimeric monoclonal antibody Bavituximab (111). Bavituximab was first modified with N-succinimidyl S-acetylthioacetate (SATA) using a commercial kit. Direct labeling with radio-arsenic was performed after deprotection of the sulfhydryl groups using hydroxylamine [Figure 2]. The resultant radioimmunoconjugate demonstrated good stability over 72 hours during incubation in fetal bovine serum as determined by size-exclusion HPLC (111).
Figure 2.
Representative scheme of radiolabeling intact antibodies with radio-arsenic using SATA approach.
Biological studies with 72As labeled antibodies
Bavituximab was labeled with 74As (t1/2 = 427 h, 30 % positron emission) as a surrogate for 72As for a proof in principle study and evaluated for PET imaging in rats bearing Dunning prostate R3327-AT1 tumors (111). Tumors were clearly visualized after 48 hours with injection of 10 MBq of the radioimmunoconjugate with the highest tumor to background ratio being achieved at 72 hours. Localization of the radio-arsenic labeled immunoconjugate was confirmed by autoradiography. As a proof of principle, this study demonstrated successful use of radio-arsenic in PET radioimmunoimaging. However, one might argue that 72As is a better candidate for radioimmunoimaging due to its more appropriate half-life and higher positron abundance. 72As has been used in humans to study environmental toxicity caused by high doses of natural arsenic (112).
Challenges
Production of 72As is limited to a few centers around the world and therefore the issue of a steady supply of clinical grade 72As remains unclear. There have been efforts by the United States Department of Energy to assist in development of 72As generators; however, the availability of a commercial generator seems unlikely in near future. Another drawback of 72As is its association with a relatively high radiation dose which could restrict certain applications particularly when hematologic toxicity is a concern since the uncomplexed arsenic localizes in red blood cells (88).
Copper-64
Copper belongs to the Group IB of the periodic table and has an atomic radius of 1.6 Å, and an electronegativity of 1.90. 64Cu has been a radionuclide of considerable interest for PET imaging due to its attractive physical and chemical properties. As compared to 18F, 64Cu has a longer half-life of 12.7 h and decays to stable 64Ni and 64Zn daughters. Besides the positron emission, 64Cu also emits Auger electrons and has a β−-emission, and therefore could potentially be used for radioimmunotherapy (113). Imaging characteristics as a result of optimum positron energy (0.66 MeV) and no high-energy γ-emission are one of the biggest factors that make 64Cu an attractive candidate for PET imaging [Table 1]. The image quality and spatial resolution are equivalent to 18F and require no major adjustments in data processing and analysis (114).
Production of 64Cu
64Cu can be produced in numerous ways. Initially, 64Cu was prepared by use of the 64Zn(n,p)64Cu reaction, but due to low yields and high contamination of 67Cu, the 64Ni(p,n)64Cu reaction was later employed to produce therapeutic quantities of 64Cu (115–117). 64Cu can also be produced through the 68Zn(p,αn)64Cu reaction using a high-energy instrument (118–120). 68Zn(p,αn)64Cu reaction gives a good yield, but the radionuclidic purity is as low as 75% and the final product contains around 25% of 67Cu as shown in Table 2 (118, 120). High yield production of good quality 64Cu has greatly improved over the past couple of years and this radionuclide is now commercially available from several sources such as Trace Life Science, Isotrace Technologies, MDS Nordion, and IBA Molecular.
Radiolabeling of antibodies with 64Cu
Copper can exist in two primary oxidation states, Cu(I) and Cu(II). It has been demonstrated that kinetic inertness is more important than thermodynamic stability of Cu(II) complexes as Cu(II) complexes of EDTA and DTPA rapidly dissociated in human serum and a Cu(II)-albumin complex was formed (121)
To improve the in vivo stability, antibodies have been labeled with 64Cu using various bifunctional chelating agents [Figure 3] such as derivatives of 1,4,8,11-tetraazacyclotetradecane (cyclam) (122), 1,4,8,11-tetraazacyclotetradecane-N, N′,N″,N‴-tetraacetic acid (TETA) (123, 124), and DOTA (125). However, all these bifunctional chelates and their derivatives were unsuccessful in preventing metabolic processing of the 64Cu complex, presumably a factor of the redox chemistry of Cu(II), and as a result elevated liver uptake was observed. To overcome this in vivo instability of Cu(II) complexes, cross-bridged tetraamine chelating agents are currently being developed and evaluated (126) These cross-bridged chelating agents provided better geometric coordination and were inert in aqueous solutions (126). Most application of these chelating agents has been for labeling peptides (127). To our best knowledge, intact antibodies have not been labeled with 64Cu using cross-bridged tetraamine chelating agents as yet due to required harsh radiolabeling conditions utilizing high temperature and acidic pH that are not conducive for labeling heat and acid sensitive antibodies. To overcome these problems, a novel chelating agent SarAr: 1-N-(4-aminobenzyl)-3,6,10,13,16,19-hexaazabicyclo[6.6.6]eicosane-1,8-diamine conjugated to anti-GD2 monoclonal murine antibody 14.G2a and the corresponding chimeric monoclonal antibody ch14.8 were evaluated recently (128). SarAr is based on the macrobicyclic sarcophagine cage structure and was modified to contain the reactive aminobenzyl group for conjugation with the antibody. At pH 5.0and room temperature conditions, the SarAr chelating agent forms a stable complex with Cu(II) within the multiple macrocyclic rings comprising the sarcophagine cage structure. Therefore, radiolabeling of SarAr-mAb was performed at less harsh conditions (128). The resulting radioimmunoconjugate was shown to be stable in presence of liver proteins known to transchelate with 64Cu (128). Alternately, attempts have been made to radiolabel antibodies with 64Cu using porphyrin based chelating agents (129).
Figure 3.
Selected chelating agents used for radiolabeling intact antibodies with 64Cu.
Biological studies with 64Cu labeled antibodies
Numerous studies using 64Cu labeled antibodies have been reported for PET radioimmunoimaging and radioimmunotherapy. Clinically approved antibodies such as Cetuximab and Abegrin have been labeled with 64Cu for PET imaging in pre-clinical models (130–132). 64Cu-DOTA-cetuximab was evaluated in mice bearing EGFR positive and EGFR negative tumors for quantitative PET imaging (132). In this study, 64Cu-DOTA-cetuximab showed prominent uptake in EGFR-expressing tumors, but low accumulation in EGFR-negative tumors. A good linear correlation between the calculated %ID/g values as measured by PET imaging and the EGFR expression level as measured by western blot was exhibited (132). Subcutaneous xenografts and peritoneal metastases were clearly visualized by PET imaging with an anti-L1-CAM antibody chCE7 labeled with 64Cu in mice bearing L1-CAM tumors (133, 134). However, this PET study also revealed that radio-metabolites of the copper immunoconjugate non-specifically localized in the lymph nodes through the reticuloendothelial system which could be critical in imaging lymph node metastasis (134). Recently, novel approaches in terms of chelating agents and linkers have been used to decrease the liver uptake and increase the target/background ratio (128, 135). The evaluation of 64Cu-SarAr-14.G2a demonstrated that the use of a better chelating agent could greatly improve the tumor/background ratio as MicroPET imaging confirmed significant uptake of the radioimmunoconjugate in GD-2-positive tumors, with minimal uptake in GD-2-negative tumors and non-target tissues of interest such as the liver (128). Another use of 64Cu labeled antibodies was to serve as a PET surrogate for 67Cu labeled antibodies used for radioimmunotherapy (13). On comparison with 67Cu, it was found that the 64Cu labeled antibody alone reduced tumor growth, however, a much higher dose (5 times higher) of 64Cu was required to produce effects comparable to 67Cu and the therapy with 64Cu was largely ineffective in treating larger tumors (136). To date, there has been one clinical study reported that utilized 64Cu-TETA-1A3 to evaluate 36 patients with suspected primary or advanced colorectal cancer (137). In this study, all patients had CT scans and PET scans 4 to 36 hours after being injected with 370 MBq of 64Cu-TETA-1A3 directed towards lipid antigens present in human colon carcinoma. The positive predictive value of 64Cu-TETA-1A3 PET radioimmunoimaging was excellent, ranging from 89% (40/45) to 96% (43/45) and detected 11 new occult tumor sites including 9 small abdominopelvic foci less than 2.0 cm in diameter that were previously not detected by CT or MRI. This study demonstrated that PET radioimmunoimaging with 64Cu may have important applications in clinical oncology, particularly for detecting smaller lesions undetected by CT or MRI.
Challenges
Excellent results have been obtained while imaging intact antibodies with 64Cu, but 64Cu seems to be a more suitable candidate for imaging when applied to use antibody fragments, diabodies, and other engineered targeting vectors which have faster localization and clearance (124, 132). Copper is a constituent of many metalloenzymes related to cellular respiratory oxidation process (88) and therefore, the release and accumulation of 64Cu from the most traditional radioimmunoconjugates in the organs such as liver remains a concern in spite of efforts to reduce the non-specific uptake by developing better chelating agents and linkers.
Yttrium-86
Yttrium belongs to Group IIIB of the periodic table. Yttrium has an atomic radius of 2.3 Å and electronegativity of 1.22. The primary purpose for exploring the viability of 86Y for PET imaging was to use it as an imaging and dosimetry surrogate for the therapeutic radionuclide 90Y which has no imageable emission. 86Y is an attractive candidate for studying 90Y due its half-life (14. 2 h) which allows imaging over several days.
Production of 86Y
86Y can be produced on a medical cyclotron by irradiating SrCO3 or SrO with 2–6 microA of beam current for <4 hours applying the 86Sr(p,n)86Y reaction. As compared to SrCO3, SrO is a superior target as a SrO target can withstand at least a 6 microA of beam current, a significant improvement over a maximum of 2 microA on the SrCO3 target resulting in yields that are almost doubled with minimum contaminants (138). At our own institution, we have developed a simple and efficient semi-automated method to purify 86Y which does not compromise the biological properties of the 86Y labeled antibody (139, 140).
Radiolabeling of antibodies with 86Y
In physiological conditions, the primary oxidation state of yttrium is Y(III). Chelation chemistry is well established for Y(III) radionuclides and several bifunctional derivatives of both DOTA and DTPA [Figure 4] have demonstrated their utility in this arena (95). The majority of the chelation chemistry for yttrium labeled antibodies was carried out with 90Y and 88Y. In 1976, 90Y-DTPA was used for radionuclide therapy of brain tumors in patients (141), and since then cyclic anhydride-DTPA was used to conjugate to antibodies for radiolabeling with 90Y (142). However, when 90Y-DTPA antibodies were evaluated in vivo, 90Y was released from that DTPA and deposited in the bones causing radiation toxicity (143, 144). In order to improve the in vivo stability of the radioimmunoconjugate, backbone substituted DTPA derivatives such as p-isothiocyanatobenzyl- diethylenetriaminepentaacetic acid (SCN-Bz-DTPA) and a methyl derivative of p-isothiocyanatobenzyl-diethylenetriaminepentaacetic acid (SCN-Bz-Mx-DTPA also known as 1B4M-DTPA) (143–145) were evaluated. The 4-isothiocyanatobenzyl group (SCN-Bz) substituted onto the carbon backbone of DTPA for use in linkage to the antibody and the methyl groups were incorporated onto the backbone to sterically hinder the release of radiometal from the chelate. Comparative biological studies in mice demonstrated the superiority of backbone substituted DTPA radioimmunoconjugates over cyclic anhydride DTPA radioimmunoconjugates (143, 144). With the better understanding of the coordination chemistry and geometry of DTPA derivatives, a cyclohexyl derivative of DTPA was developed which demonstrated excellent stability in vitro and in vivo (146, 147). Interestingly, stereochemistry of CHX-DTPA played a major role in in vivo stability of 90Y/88Y radioimmunoconjugate and subsequent bone uptake (146, 148). The single enantiomeric [(R)-2-amino-3-(4-isothiocyanatophenyl)propyl]-trans-(S,S)-cyclohexane-1,2-diamine-pentaacetic acid (CHX-A″-DTPA) radioimmunoconjugate was found to be more stable as compared to the corresponding diastereomeric pair of CHX-B-DTPA radioimmunoconjugates (148). After 7 days post injection, bone uptake of the 88Y labeled CHX-B′-DTPA or CHX-B″-DTPA radioimmunoconjugates was 5 times higher than that of the 88Y labeled CHX-A″-DTPA radioimmunoconjugate (148). In order to further improve the stability, DOTA derivatives such as SCN-Bz-DOTA were explored for radiolabeling antibodies with 90Y (149, 150). In a comparative study, it was found that DOTA conjugated antibody was better than DTPA conjugated antibody in terms of stability (150). Since then numerous pre-clinical and clinical studies have been published reporting DTPA and DOTA based chelating agents for 90Y radioimmunotherapy and thus extrapolating to 86Y for PET imaging (151–153).
Figure 4.
Selected chelating agents used for radiolabeling intact antibodies with 86Y/90Y.
Biological studies with 86Y labeled antibodies
86Y labeled antibodies were most recently evaluated to perform dosimetry for 90Y labeled antibodies used for radioimmunotherapy as they were expected to be superior to 111In labeled antibodies in terms of similarities in both chemical and biological behavior (153). Numerous studies have been reported with 86Y labeled antibodies including clinically approved trastuzumab (Herceptin) (152, 154) and cetuximab (Erbitux) (154). In a recent study, 86Y-CHX-A″-DTPA-trastuzumab was evaluated in mice bearing intraperitoneal SKOV3 ovarian tumors (152). In this study, PET and MRI studies were performed to assess the therapeutic potential of 90Y-CHX-A″-DTPA-trastuzumab using 86Y-CHX-A″-DTPA-trastuzumab as an imaging surrogate for dosimetry. Intraperitoneal tumors were visualized by PET imaging 2 days after injecting 86Y-CHX-A″-DTPA-trastuzumab and the tumor localization was confirmed by MRI studies. 86Y-CHX-A″-DTPA-trastuzumab localized to sites of disease with minimal normal organ uptake (152). In the study performed at our institution, we could clearly visualize LS-174T colon carcinoma xenografts in mice after 2 days of injecting 86Y-CHX-A″-DTPA trastuzumab (tumor uptake was greater than 30% ID/g) (154). In the same study, the HER1-targeted 86Y-CHX-A″-DTPA cetuximab was evaluated in mice bearing LS-174T colon carcinoma, SKOV3 ovarian carcinoma and DU-145 prostate carcinoma (154). All the tumors were clearly visualized at 1 day post-injection with the tumor uptake ranging from 20–30% ID/g.
Clinical use of 86Y labeled agent used for dosimetry 90Y labeled has been illustrated by the use of 86Y-DOTA-Phe1-Tyr3-octreotide (SMT-487) for dosimetry of 90Y-SMT-487 in patients with metastatic carcinoid tumors (155). In this study, the maximum dose that could be delivered to the tumor without exceeding the 23-Gy cut-off dose to kidneys was successfully calculated using PET imaging, and therefore presumably reduced the incidence of potential renal toxicity caused by 90Y-SMT-487 therapy.
Challenges
86Y is commercially available (Isotrace Technology) in a limited capacity and is also only available at centers capable of producing 86Y on a regular basis. The high positron energy of 3.14 MeV (34% abundance) and addition γ-emission of 1.08 MeV (83 % abundance) significantly affects the spatial resolution and image quality respectively [Table 1]. Uncomplexed 86Y localizes in the bone and therefore can cause radiation toxicity in the events of “leakage” from the immunoconjugate and thus may be not a suitable candidate to image bone metastasis.
Zirconium-89
Zirconium belongs to Group IVB of the periodic table, has an atomic radius of 2.2 Å, and an electronegativity of 1.33. 89Zr (t1/2= 78.4 h) decays to an ultra short-lived daughter 89mY (t1/2= 16 s) with a 23% β+ emission and 77% electron capture [Table 1]. Due to its appropriate half-life and imaging characteristics, 89Zr labeled antibodies are currently being evaluated in the clinic as PET surrogates for both 90Y and 177Lu therapy studies despite the possibility of using the elementally matched 86Y for the former and that the latter possesses an imagable γ-emission (156).
Production of 89Zr
89Zr can be produced through the 89Y(p,n)89Zr reaction in which inexpensive natural yttrium foil is irradiated with a 14–14.5 MeV proton beam and the target is separated by ion-exchange chromatography or solvent extraction (26, 157, 158). Alternatively, 89Zr can also be produced through the 89Y(d,2n)89Zr reaction (159). A relatively high yield of 43 MBq/μAh is obtained with the 89Y(p,n)89Zr reaction with less than 0.2% impurity of 88Zr (t1/2=85 d and daughter 88Y; t1/2=105 d) [Table 2].
Radiolabeling of antibodies with 89Zr
Over the past 20 years attempts have been made to label proteins and antibodies with 89Zr using DTPA derivatives and porphyrins as chelating agents (160, 161). Proteins and antibodies were labeled with 89Zr using desferal (Df) conjugates (162, 163). Briefly, incorporation of Df-groups onto monoclonal antibodies was performed in a two-step procedure. In the first step, maleimide groups were incorporated into the protein with the assistance of succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC). In the second step, the thioester of Df formed by conjugation to SATA was further converted to a free thiol with hydroxylamine to facilitate reaction with the maleimide groups of the antibody with subsequent radiolabeling with 89Zr. The final purified product was obtained through gel filtration (162, 163). This protocol was further modified by formation of N-succinyldesferrioxamine B of Df (N-SucDf) as an intermediate (164). In this method, the hydroxamate groups of N-sucDf were temporarily blocked with formation of the Fe(III) complex. The N-sucDf-Fe complex was then esterified and coupled to antibodies. Thereafter, Fe(III) was removed by reduction to Fe(II) and transchelation to EDTA, mAb-N-sucDf is then radiolabeled with 89Zr at pH 7.2-7.4 with a 30 minute incubation at room temperature (164).
Biological studies with 89Zr labeled antibodies
Considering the success of 90Y-labeled mAbs in clinical radioimmunotherapy (12, 151, 165), 86Y-labeled mAbs are currently being developed in pre-clinical models for clinical dosimetry purposes (139, 152, 153). However, the half-life of 90Y is nearly 4 times greater than that of 86Y and therefore predicting 90Y dosimetry of slowly clearing antibody may not be feasible or even appropriate. Due to these reasons, 89Zr was explored as one of the possible radionuclides that could be used for dosimetry analysis of 90Y-labeled mAbs (166). Clinically used antibodies such as HER1-targeted cetuximab (156), VEGF-targeted bevacizumab (167) and CD-20-targeted ibritumomab tiuxetan have been labeled with 89Zr-Df conjugate (168). All the above studies demonstrated the use of 89Zr labeled antibodies in quantitative PET imaging; additionally, the tumor localization of the 89Zr-mAb was similar to that of the 111In/90Y-mAb.
Due to the feasibility in production and purification of 89Zr labeled mAb, clinical studies were performed with CD44V6-specific chimeric mAb, U36 labeled with 89Zr (169). In this study, 89Zr-U36 was evaluated in patients with squamous cell carcinoma of the head and neck (HNSCC), who were at high risk of having neck lymph node metastases. All primary tumors (n = 17) as well as lymph node metastases in 18 of 25 positive levels (sensitivity 72%) and in 11 of 15 positive sides (sensitivity 73%) were detected using PET radioimmunoimaging (169). Interpretation of PET was correct in 112 of 121 operated levels (accuracy 93%) and in 19 of 25 operated sides (accuracy 76%). For CT/MRI, sensitivities of 60% and 73% and accuracies of 90% and 80% were found per level and side, respectively. 89Zr-immunoPET demonstrated a sensitivity of 85% and accuracy of 95% as compared to sensitivity of 62 % and accuracy of 88% for18F-FDG-PET (169). Thus, this study demonstrated the utility and advantages of 89Zr PET radioimmunoimaging in clinical setting in comparison with CT/MRI and 18F-FDG (169). Similarly, all known tumor lesions previously identified by 18F-FDG were successfully imaged by 89Zr-Df-Zevalin in a pilot PET imaging study in a patient with Non-Hodgkin’s Lymphoma (168)
Challenges
Uncomplexed 89Zr localizes in the bone and therefore can deliver high radiation dose to the bone marrow, therefore the need for stable chelation chemistry remains an important consideration with this element (88). In spite of the recent success of 89Zr PET imaging, clinical grade 89Zr is not commercially available yet (although, 89Zr can be obtained from IBA Molecular on request). Another major disadvantage is the currently used 89Zr-mAb production scheme. The currently used conjugation and radiolabeling schemes are often tedious, time-consuming and involve numerous steps that increase the margin for error for the cGMP (current Good Manufacturing Practice) production. In the currently used procedure, the Fe(III) complex is first formed with desferal which is then activated for conjugation through extension of the terminal amine with succinic anhydride followed by the conversion of the formed carboxylate into an active ester. After conjugation of the antibody, the Fe(III) is then reduced and displaced with 89Zr (164). Alternate, simpler conjugation and radiolabeling schemes that can be successfully carried out at local radiopharmacies are warranted to bolster theincreased appeal of 89Zr PET radioimmunoimaging. One of the suggested applications of 89Zr has been to use it as a surrogate for 90Y and 131I labeled antibodies, however, 89Zr labeled antibodies, while close to, also do not exactly match the biodistribution and pharmacokinetics of 90Y and 131I labeled antibodies (168, 170).
Opportunities and Challenges
Production
The reliable large-scale availability of the above desired longer-lived positron emitting radionuclides in suitable quality for clinical applications is critical for potential clinical translation. Many of the above described radionuclides such as 64Cu, 76Br, 86Y and 72As can be produced in good yields at low-energy medical cyclotrons (26). Most of the longer-lived positron-emitting radionuclides are currently produced by employing low-energy nuclear reactions such as (p, n) and (d, n) which may require isotopically enriched targets which are usually expensive. Alternatively, a higher-energy dual-particle cyclotron or a multi-particle intermediate-energy cyclotron could be used for production of some these radionuclides employing nuclear reactions such as (p, xn) and (3He, xn). Two of the most important criteria in medical application of positron-emitters are quality and yield, and therefore the target material is critical. It is absolutely essential to produce target material that is economical, may be efficiently recovered for reuse, can withstand high-energy beam fluxes of charged particles, and most importantly, also be easily isolated from the product to give high-purity final product. One such example that meets these criteria has been the production of 64Cu. Originally, 64Cu was produced through the 64Zn(n,p)64Cu reaction with resulting in low yields and a high contamination of 67Cu, unless highly enriched pure 64Zn was used. The low yields compounded with contamination of the β−-emitting therapeutic radionuclide 67Cu were impediments to routine large-scale clinical applications of 64Cu for PET imaging. As an alternative, the 64Ni(p,n)64Cu reaction was employed to obtain higher yields and better quality 64Cu (115, 116). However, the biggest drawback of this method remains the cost of the highly enriched 64Ni target material which can be a hindrance in large-scale production of 64Cu at multiple centers across the world. With better understanding and characterization of cross-section data, integral yields and chemical processing methods, it is possible to obtain good quality positron-emitters in large yields using a medical cyclotron that can be potentially used for radioimmunoimaging. Most commonly used production routes of selected longer-lived positron emitters for radioimmunoimaging with their corresponding yields and radionuclidic impurities are listed in Table 2.
Imaging characteristics
One of the major factors impacting image quality is the additional γ-emissions and the energy of the positron which can contribute to the background noise and the loss of intrinsic spatial resolution respectively. Some of the longer-lived positron-emitting radionuclides such as 76Br, 72As, 124I and 66Ga have high-energy positron emissions which result in loss of intrinsic spatial resolution and poor signal to noise ratios [Table 1]. In clinical settings, both radiation dosimetry and chemistry as a result of inadequate specific activity and stability may limit the amount of radioactivity injected, thereby further degrading image quality. Loss of spatial resolution could also limit the use of radioimmunoconjugates labeled with some of the described longer-lived positron-emitting radionuclides for the detection of metastasis and smaller lesions which could be critical for their use in assessing or predicting clinical outcomes. Successful attempts have been made to improve the image characteristics of high-energy positron emitters by developing methods to correct for scatter fractions and spurious coincidences (171–173). Image reconstruction algorithms have been developed which include models of the statistical nature of nuclear decays and tomography responses which improve the spatial resolution, and at the same time reduce the image noise (114, 171–173). Further advances in instrumentation and image processing may additionally improve spatial resolution and the ability to perform more accurate quantitative analysis. However, implementing these improvements in the clinic may be complex as new instruments and imaging software will require FDA approval.
Potential therapeutic applications and radiation toxicity
Tumor therapy using antibodies carrying radioactivity has been reported now for over 50 years (174, 175). In 1966, McCradle and co-workers reported the use of 131I labeled antibody to human fibrinogen as a clinical diagnostic and therapeutic agent (8). In this study, 50 patients with reported neoplasm were imaged and a high percentage of patients showed positive scans for metastases to the brain which was a remarkable achievement during that period when targeted non-invasive radioimmunoimaging was still in its infancy. This study demonstrated the value of molecular imaging with radiolabeled antibodies for the assessment of primary and metastatic lesions (8). In this same study, two previously imaged patients were treated with higher doses of 131I labeled antibody to human fibrinogen resulting in regression of their lesions (8). Since that time numerous studies have been reported demonstrating the utility of antibodies labeled with radionuclides such as 111In, 177Lu and 64Cu which could be used for diagnosis as well as therapy (4, 176). For better clinical acceptance, cost-benefit effectiveness and numerous other reasons including the need of surrogate radionuclide, a radiolabeled antibody that could be used as a diagnostic as well as therapeutic becomes a desirable combination.
One of the major concerns in radioimmunotherapy is the non-uniform absorbed dose distribution in the tumor resulting in less effective therapy in spite of dose escalation (177). One possible solution might be use of longer-lived radionuclides with longer emission ranges which may increase mean absorbed dose to the tumor as they are more likely to penetrate into the tumor before decaying in the body than shorter-lived radionuclides (178). Longer-lived positron-emitting radionuclides can deliver high absorbed doses to the target site as the doses delivered by positrons are as high as negatrons of the same energy. Considering the success of negatron-emitting radioimmunotherapy in pre-clinical and clinical studies, longer-lived positron-emitting radioimmunoconjugates may potentially be used for therapeutic applications. The longer half-life may minimize the effects of non-uniformity up to an extent. However, the choice of radionuclide will be based upon the function of biological and pharmacokinetic properties of the targeting antibody (normal organs uptake and clearance), disease state, physical half-life of the radionuclide, abundance and energy of the positron emission and additional γ-rays. For example, assuming a biological half-life of 100 hours and whole-body uniform distribution in an adult human, based on the values given in the International Commission on Radiological Protection (ICRP) publications, the effective dose from 52Mn (t1/2= 134.2 h) will be approximately 0.9–1.1 mSv/MBq as compared to 0.02–0.04 mSv/MBq from 64Cu (t1/2= 12.7 h) and 0.2–0.3 mSv/MBq from 76Br (t1/2= 16.2 h) (159). Another important parameter in terms of therapeutic applications of longer-lived positron emitters is the positron abundance and the dose rate; a dose rate of 2 – 3 cGy/hr has been suggested as being required to overcome the proliferation of most types of tumor cells (179). Therefore, the portion of the total dose that is delivered below this rate is essentially unnecessary and can potentially pose a hazard. Theoretically, high specific activity longer-lived positron-emitting radioimmunoconjugates could be used for therapy; however toxicity and dose to the normal organs may pose a few challenges and will depend on whether the radioimmunoconjugate is delivered systemically or locally.
Economics, regulatory affairs and clinical acceptance
Currently, the use of PET in the clinical setting revolves around 18F-FDG, which has great clinical acceptance as it can be used to image most metabolically active tumors and lesions irrespective of any expression of cell surface molecular targets such as EGFR and HER2/μ. This characteristics of 18F-FDG can also be disadvantageous too as it does not provide information regarding the status of cell surface targets which could be essential for patients about to undergo immunotherapy or radioimmunotherapy. Under those circumstances, PET radioimmunoimaging could play a critical role in risk stratification. In the past year, sales of Herceptin (trastuzumab), Avastin (Bevacizumab), Rituxan (Rituximab) and Erbitux (Cetuximab) each all exceeded one billion U.S. dollars (180). Based on previous results, not all patients may respond to immunotherapy due to low-density of target expression or changes in the target levels. In all likelihood, not all patients undergo some form of diagnostic scan to evaluate the status of target in the progression of illness. PET radioimmunoimaging could serve as one of the potential tools for screening patients that may or may not respond to therapy, thereby acting as a cost containment tool. Contrary to that positive aspect, a sizeable financial investment and commitment is required to develop a clinically FDA approved radiopharmaceutical. In order to recover the cost spent in developing the PET radioimmunoconjugate, sales and use volumes have to be significantly high which will be directly related to the clinical acceptance of PET radioimmunoimaging.
The number of radiopharmaceuticals approved by the US FDA in the past decade has been limited due to more scrutiny and a tedious approval process. Radiation toxicity and required pre-approval studies could potentially be a major obstacle for the approval of radioimmunoconjugates labeled with longer-lived positron emitters. Thus, one can question the rationale behind the objective of developing a novel PET radioimmunoconjugate considering the level of required investment and resources versus failure to acquire approval of the agent by the FDA.
Other regulatory barriers include waste disposal of large quantities of longer-lived high energy positron emitters, decontamination, occupational hazards, and the radiation exposure to normal population by out-patients undergoing high-dose longer-lived positron-emitting radionuclide protocols.
Lack of standards for calibration, decay characteristics
A recent study has highlighted the differences in published values of decay data and energies of some of the described radionuclides (119). These differences in values could impact the overall biological and dosimetry results as the same amount of radioactivity quantified from dose calibrator and PET image could vary from institution to institution depending of the values used. Therefore, there is a need to establish standards for calibration of instruments for these longer-lived positron emitters to avoid variances which could result in errors.
Future perspective
It is clear from the selected examples cited in this report that efforts are continuing to being made to develop radioimmunoconjugates labeled with longer-lived positron-emitting radionuclides for PET imaging. The superiority of PET in radioimmunoimaging was demonstrated by a recent clinical study in which a comparison was made between 131I-G250, 124I-cG250 and 18F-FDG for detection of metastatic renal-cell carcinoma (53, 181). 131I-cG250 was able to detect 30% of metastatic lesions as compared to 69% for 18F-FDG (181); however, the results were far better when 124I-cG250 was used as the sensitivity of detection of tumor was 94 % (53, 181). When compared to 124I-cG250, 131I-cG250 did not provide the same resolution, contrast, or the potential for quantification as 124I-cG250 (53). Considering the advantages of PET radioimmunoimaging, further efforts are warranted to improve PET instrumentation and image processing, increase production of clinical grade high-yield longer-lived positron emitters, and continue to create efficient radiolabeling methods to produce high-yield biologically active stable radioimmunoconjugates in the least amount of time and variability. And, lastly, but perhaps the most critical criterion for successful clinical translation, continued efforts have to be applied towards making wise selections of the most appropriate antibody matched with the most appropriate radionuclide for the desired application within an appropriate patient population. However, a few questions still remain unanswered: how many positron-emitting radionuclides do we really need to evaluate to “fit the bill” and to be able to ask and answer questions related to radioimmunoimaging? What are the alternatives in case of unexpected unreliable availability of the radionuclide? Is it worth the capital investments related to PET imaging if techniques such as biopsies and CT imaging are going to remain the mainstay of cancer diagnosis? Can future generation SPECT systems be as good as currently used PET (182, 183)? If so, then why develop newer positron-emitting products if 111In based SPECT can offer precise quantitation in future? And finally, should be there be a common set of criteria and guidelines for choosing the radionuclide?
In conclusion, we believe that radioimmunoimaging with longer-lived positron-emitting radionuclides holds significant promise, but several scientific and non-scientific challenges have to be conquered before realizing the true potentials of PET radioimmunoimaging with intact antibodies.
Acknowledgments
This report was supported by the Intramural Research Program of the NIH, NCI, Center for Cancer Research and the United States Department of Health and Human Services. Gratitude is expressed to Mike Slackenerny for constant encouragement and support.
Footnotes
Authors declare no conflict of interests.
Literature Cited
- 1.Cattell HW. Application of the X-Rays to Surgery. Science. 1896;3:344–346. doi: 10.1126/science.3.62.344. [DOI] [PubMed] [Google Scholar]
- 2.Wagner HN, Jr, Bardfeld PA. Evaluation of structure and function of spleen with radioactive tracers. JAMA. 1967;199:202–6. [PubMed] [Google Scholar]
- 3.Wagner HN., Jr The current status of nuclear medicine. JAMA. 1963;183:500–3. doi: 10.1001/jama.1963.63700060052035. [DOI] [PubMed] [Google Scholar]
- 4.Boswell CA, Brechbiel MW. Development of radioimmunotherapeutic and diagnostic antibodies: an inside-out view. Nucl Med Biol. 2007;34:757–78. doi: 10.1016/j.nucmedbio.2007.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Khaw BA. Antibodies for molecular imaging in the cardiovascular system. J Nucl Cardiol. 2005;12:591–604. doi: 10.1016/j.nuclcard.2005.07.001. [DOI] [PubMed] [Google Scholar]
- 6.Melcher LR, Masouredis SP. The in vivo stability of the I131 protein label of rabbit antibody in guinea pigs as determined by the quantitative precipitin reaction. J Immunol. 1951;67:393–402. [PubMed] [Google Scholar]
- 7.Pressman D. The zone of localization of antibodies; the specific localization of antibodies to rat kidney. Cancer. 1949;2:697–700. doi: 10.1002/1097-0142(194907)2:4<697::aid-cncr2820020416>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
- 8.McCardle RJ, Harper PV, Spar IL, Bale WF, Andros G, Jiminez F. Studies with iodine-131-labeled antibody to human fibrinogen for diagnosis and therapy of tumors. J Nucl Med. 1966;7:837–47. [PubMed] [Google Scholar]
- 9.Goldenberg DM, DeLand F, Kim E, Bennett S, Primus FJ, van Nagell JR, Jr, Estes N, DeSimone P, Rayburn P. Use of radiolabeled antibodies to carcinoembryonic antigen for the detection and localization of diverse cancers by external photoscanning. N Engl J Med. 1978;298:1384–6. doi: 10.1056/NEJM197806222982503. [DOI] [PubMed] [Google Scholar]
- 10.Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature. 1975;256:495–7. doi: 10.1038/256495a0. [DOI] [PubMed] [Google Scholar]
- 11.Goldenberg DM, Kim EE, Deland F, Spremulli E, Nelson MO, Gockerman JP, Primus FJ, Corgan RL, Alpert E. Clinical studies on the radioimmunodetection of tumors containing alpha-fetoprotein. Cancer. 1980;45:2500–5. doi: 10.1002/1097-0142(19800515)45:10<2500::aid-cncr2820451006>3.0.co;2-j. [DOI] [PubMed] [Google Scholar]
- 12.O’Donnell RT, DeNardo SJ, Yuan A, Shen S, Richman CM, Lara PN, Griffith IJ, Goldstein DS, Kukis DL, Martinez GS, Mirick GR, DeNardo GL, Meyers FJ. Radioimmunotherapy with (111)In/(90)Y-2IT-BAD-m170 for metastatic prostate cancer. Clin Cancer Res. 2001;7:1561–8. [PubMed] [Google Scholar]
- 13.DeNardo GL, Kukis DL, Shen S, DeNardo DA, Meares CF, DeNardo SJ. 67Cu-versus 131I-labeled Lym-1 antibody: comparative pharmacokinetics and dosimetry in patients with non-Hodgkin’s lymphoma. Clin Cancer Res. 1999;5:533–41. [PubMed] [Google Scholar]
- 14.Mach JP, Carrel S, Forni M, Ritschard J, Donath A, Alberto P. Tumor localization of radiolabeled antibodies against carcinoembryonic antigen in patients with carcinoma: a critical evaluation. N Engl J Med. 1980;303:5–10. doi: 10.1056/NEJM198007033030102. [DOI] [PubMed] [Google Scholar]
- 15.Phelps ME. PET: the merging of biology and imaging into molecular imaging. J Nucl Med. 2000;41:661–81. [PubMed] [Google Scholar]
- 16.Otsuka FL, Welch MJ, Kilbourn MR, Dence CS, Dilley WG, Wells SA., Jr Antibody fragments labeled with fluorine-18 and gallium-68: in vivo comparison with indium-111 and iodine-125-labeled fragments. Int J Rad Appl Instrum B. 1991;18:813–6. doi: 10.1016/0883-2897(91)90023-e. [DOI] [PubMed] [Google Scholar]
- 17.Garg PK, Garg S, Bigner DD, Zalutsky MR. Localization of fluorine-18-labeled Mel-14 monoclonal antibody F(ab′)2 fragment in a subcutaneous xenograft model. Cancer Res. 1992;52:5054–60. [PubMed] [Google Scholar]
- 18.Cai W, Olafsen T, Zhang X, Cao Q, Gambhir SS, Williams LE, Wu AM, Chen X. PET imaging of colorectal cancer in xenograft-bearing mice by use of an 18F-labeled T84.66 anti-carcinoembryonic antigen diabody. J Nucl Med. 2007;48:304–10. [PubMed] [Google Scholar]
- 19.Kramer-Marek G, Kiesewetter DO, Martiniova L, Jagoda E, Lee SB, Capala J. [(18)F]FBEM-Z(HER2:342)-Affibody molecule-a new molecular tracer for in vivo monitoring of HER2 expression by positron emission tomography. Eur J Nucl Med Mol Imaging. 2008;35:1008–18. doi: 10.1007/s00259-007-0658-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Vaidyanathan G, Bigner DD, Zalutsky MR. Fluorine-18-labeled monoclonal antibody fragments: a potential approach for combining radioimmunoscintigraphy and positron emission tomography. J Nucl Med. 1992;33:1535–41. [PubMed] [Google Scholar]
- 21.Snook DE, Rowlinson-Busza G, Sharma HL, Epenetos AA. Preparation and in vivo study of 124I-labelled monoclonal antibody H17E2 in a human tumour xenograft model. A prelude to positron emission tomography (PET) Br J Cancer Suppl. 1990;10:89–91. [PMC free article] [PubMed] [Google Scholar]
- 22.Pentlow KS, Graham MC, Lambrecht RM, Cheung NK, Larson SM. Quantitative imaging of I-124 using positron emission tomography with applications to radioimmunodiagnosis and radioimmunotherapy. Med Phys. 1991;18:357–66. doi: 10.1118/1.596728. [DOI] [PubMed] [Google Scholar]
- 23.Bakir MA, Eccles S, Babich JW, Aftab N, Styles J, Dean CJ, Lambrecht RM, Ott RJ. c-erbB2 protein overexpression in breast cancer as a target for PET using iodine-124-labeled monoclonal antibodies. J Nucl Med. 1992;33:2154–60. [PubMed] [Google Scholar]
- 24.Hohn A, Nortier FM, Scholten B, van der Walt TN, Coenen HH, Qaim SM. Excitation functions of 125Te(p, xn)-reactions from their respective thresholds up to 100 MeV with special reference to the production of 124I. Appl Radiat Isot. 2001;55:149–56. doi: 10.1016/s0969-8043(00)00388-2. [DOI] [PubMed] [Google Scholar]
- 25.Sajjad M, Bars E, Nabi HA. Optimization of 124I production via 124Te(p,n)124I reaction. Appl Radiat Isot. 2006;64:965–70. doi: 10.1016/j.apradiso.2006.04.004. [DOI] [PubMed] [Google Scholar]
- 26.Tang L. Radionuclide production and yields at Washington University School of Medicine. Q J Nucl Med Mol Imaging. 2007;52:122–33. [PubMed] [Google Scholar]
- 27.Kondo K, Lambrecht RM, Wolf AP. Iodine-123 production for radiopharmaceuticals--XX excitation functions of the 124Te(p, 2n)123I and 124Te(p, n)124I reactions and the effect of target enrichment on radionuclidic purity. Int J Appl Radiat Isot. 1977;28:395–401. doi: 10.1016/0020-708x(77)90132-6. [DOI] [PubMed] [Google Scholar]
- 28.Pressman D, Hill RF, Foote FW. The Zone of Localization of Anti-Mouse-Kidney Serum as Determined by Radioautographs. Science. 1949;109:65–66. doi: 10.1126/science.109.2821.65. [DOI] [PubMed] [Google Scholar]
- 29.Yalow RS, Berson SA. Assay of plasma insulin in human subjects by immunological methods. Nature. 1959;184(Suppl 21):1648–9. doi: 10.1038/1841648b0. [DOI] [PubMed] [Google Scholar]
- 30.Hunter WM, Greenwood FC. Preparation of iodine-131 labelled human growth hormone of high specific activity. Nature. 1962;194:495–6. doi: 10.1038/194495a0. [DOI] [PubMed] [Google Scholar]
- 31.Greenwood FC, Hunter WM, Glover JS. The Preparation of I-131-Labelled Human Growth Hormone of High Specific Radioactivity. Biochem J. 1963;89:114–23. doi: 10.1042/bj0890114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Marchalonis JJ. An enzymic method for the trace iodination of immunoglobulins and other proteins. Biochem J. 1969;113:299–305. doi: 10.1042/bj1130299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.David GS. Solid state lactoperoxidase: a highly stable enzyme for simple, gentle iodination of proteins. Biochem Biophys Res Commun. 1972;48:464–71. doi: 10.1016/s0006-291x(72)80074-3. [DOI] [PubMed] [Google Scholar]
- 34.Rudinger J, Ruegg U. Appendix: Preparation of N-succinimidyl 3-(4-hydroxyphenyl)propionate. Biochem J. 1973;133:538–539. doi: 10.1042/bj1330538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bolton AE, Hunter WM. The labelling of proteins to high specific radioactivities by conjugation to a 125I-containing acylating agent. Biochem J. 1973;133:529–39. doi: 10.1042/bj1330529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Salacinski PR, McLean C, Sykes JE, Clement-Jones VV, Lowry PJ. Iodination of proteins, glycoproteins, and peptides using a solid-phase oxidizing agent, 1,3,4,6-tetrachloro-3 alpha,6 alpha-diphenyl glycoluril (Iodogen) Anal Biochem. 1981;117:136–46. doi: 10.1016/0003-2697(81)90703-x. [DOI] [PubMed] [Google Scholar]
- 37.Wilbur DS, Hadley SW, Hylarides MD, Abrams PG, Beaumier PA, Morgan AC, Reno JM, Fritzberg AR. Development of a stable radioiodinating reagent to label monoclonal antibodies for radiotherapy of cancer. J Nucl Med. 1989;30:216–26. [PubMed] [Google Scholar]
- 38.Press OW, DeSantes K, Anderson SK, Geissler F. Inhibition of catabolism of radiolabeled antibodies by tumor cells using lysosomotropic amines and carboxylic ionophores. Cancer Res. 1990;50:1243–50. [PubMed] [Google Scholar]
- 39.Geissler F, Anderson SK, Press O. Intracellular catabolism of radiolabeled anti-CD3 antibodies by leukemic T cells. Cell Immunol. 1991;137:96–110. doi: 10.1016/0008-8749(91)90060-o. [DOI] [PubMed] [Google Scholar]
- 40.Geissler F, Anderson SK, Venkatesan P, Press O. Intracellular catabolism of radiolabeled anti-mu antibodies by malignant B-cells. Cancer Res. 1992;52:2907–15. [PubMed] [Google Scholar]
- 41.Sharkey RM, Behr TM, Mattes MJ, Stein R, Griffiths GL, Shih LB, Hansen HJ, Blumenthal RD, Dunn RM, Juweid ME, Goldenberg DM. Advantage of residualizing radiolabels for an internalizing antibody against the B-cell lymphoma antigen, CD22. Cancer Immunol Immunother. 1997;44:179–88. doi: 10.1007/s002620050371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mattes MJ, Shih LB, Govindan SV, Sharkey RM, Ong GL, Xuan H, Goldenberg DM. The advantage of residualizing radiolabels for targeting B-cell lymphomas with a radiolabeled anti-CD22 monoclonal antibody. Int J Cancer. 1997;71:429–35. doi: 10.1002/(sici)1097-0215(19970502)71:3<429::aid-ijc21>3.0.co;2-9. [DOI] [PubMed] [Google Scholar]
- 43.Stein R, Goldenberg DM, Thorpe SR, Basu A, Mattes MJ. Effects of radiolabeling monoclonal antibodies with a residualizing iodine radiolabel on the accretion of radioisotope in tumors. Cancer Res. 1995;55:3132–9. [PubMed] [Google Scholar]
- 44.Stein R, Govindan SV, Mattes MJ, Chen S, Reed L, Newsome G, McBride BJ, Griffiths GL, Hansen HJ, Goldenberg DM. Improved iodine radiolabels for monoclonal antibody therapy. Cancer Res. 2003;63:111–8. [PubMed] [Google Scholar]
- 45.Wilbur DS. Radiohalogenation of proteins: an overview of radionuclides, labeling methods, and reagents for conjugate labeling. Bioconjug Chem. 1992;3:433–70. doi: 10.1021/bc00018a001. [DOI] [PubMed] [Google Scholar]
- 46.Collingridge DR, Carroll VA, Glaser M, Aboagye EO, Osman S, Hutchinson OC, Barthel H, Luthra SK, Brady F, Bicknell R, Price P, Harris AL. The development of [(124)I]iodinated-VG76e: a novel tracer for imaging vascular endothelial growth factor in vivo using positron emission tomography. Cancer Res. 2002;62:5912–9. [PubMed] [Google Scholar]
- 47.Sundaresan G, Yazaki PJ, Shively JE, Finn RD, Larson SM, Raubitschek AA, Williams LE, Chatziioannou AF, Gambhir SS, Wu AM. 124I-labeled engineered anti-CEA minibodies and diabodies allow high-contrast, antigen-specific small-animal PET imaging of xenografts in athymic mice. J Nucl Med. 2003;44:1962–9. [PMC free article] [PubMed] [Google Scholar]
- 48.Fortin MA, Salnikov AV, Nestor M, Heldin NE, Rubin K, Lundqvist H. Immuno-PET of undifferentiated thyroid carcinoma with radioiodine-labelled antibody cMAb U36: application to antibody tumour uptake studies. Eur J Nucl Med Mol Imaging. 2007;34:1376–87. doi: 10.1007/s00259-006-0346-5. [DOI] [PubMed] [Google Scholar]
- 49.Phan HT, Jager PL, Paans AM, Plukker JT, Sturkenboom MG, Sluiter WJ, Wolffenbuttel BH, Dierckx RA, Links TP. The diagnostic value of (124)I-PET in patients with differentiated thyroid cancer. Eur J Nucl Med Mol Imaging. 2008 doi: 10.1007/s00259-007-0660-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Freudenberg LS, Antoch G, Frilling A, Jentzen W, Rosenbaum SJ, Kuhl H, Bockisch A, Gorges R. Combined metabolic and morphologic imaging in thyroid carcinoma patients with elevated serum thyroglobulin and negative cervical ultrasonography: role of (124)I-PET/CT and FDG-PET. Eur J Nucl Med Mol Imaging. 2008 doi: 10.1007/s00259-007-0634-8. [DOI] [PubMed] [Google Scholar]
- 51.Kolbert KS, Pentlow KS, Pearson JR, Sheikh A, Finn RD, Humm JL, Larson SM. Prediction of absorbed dose to normal organs in thyroid cancer patients treated with 131I by use of 124I PET and 3-dimensional internal dosimetry software. J Nucl Med. 2007;48:143–9. [PubMed] [Google Scholar]
- 52.Jayson GC, Zweit J, Jackson A, Mulatero C, Julyan P, Ranson M, Broughton L, Wagstaff J, Hakannson L, Groenewegen G, Bailey J, Smith N, Hastings D, Lawrance J, Haroon H, Ward T, McGown AT, Tang M, Levitt D, Marreaud S, Lehmann FF, Herold M, Zwierzina H. Molecular imaging and biological evaluation of HuMV833 anti-VEGF antibody: implications for trial design of antiangiogenic antibodies. J Natl Cancer Inst. 2002;94:1484–93. doi: 10.1093/jnci/94.19.1484. [DOI] [PubMed] [Google Scholar]
- 53.Divgi CR, Pandit-Taskar N, Jungbluth AA, Reuter VE, Gonen M, Ruan S, Pierre C, Nagel A, Pryma DA, Humm J, Larson SM, Old LJ, Russo P. Preoperative characterisation of clear-cell renal carcinoma using iodine-124-labelled antibody chimeric G250 (124I-cG250) and PET in patients with renal masses: a phase I trial. Lancet Oncol. 2007;8:304–10. doi: 10.1016/S1470-2045(07)70044-X. [DOI] [PubMed] [Google Scholar]
- 54.Vedeneyev VIGLV, Kondrat’Yev VN, Medvedev VA. Bond Energies, ionization Potentials and electron Affinities. Edward Arnold; London: 1966. [Google Scholar]
- 55.Orlova A, Hoglund J, Lubberink M, Lebeda O, Gedda L, Lundqvist H, Tolmachev V, Sundin A. Comparative biodistribution of the radiohalogenated (Br, I and At) antibody A33. Implications for in vivo dosimetry. Cancer Biother Radiopharm. 2002;17:385–96. doi: 10.1089/108497802760363187. [DOI] [PubMed] [Google Scholar]
- 56.Rossin R, Berndorff D, Friebe M, Dinkelborg LM, Welch MJ. Small-animal PET of tumor angiogenesis using a (76)Br-labeled human recombinant antibody fragment to the ED-B domain of fibronectin. J Nucl Med. 2007;48:1172–9. doi: 10.2967/jnumed.107.040477. [DOI] [PubMed] [Google Scholar]
- 57.Bergstrom M, Lu L, Fasth KJ, Wu F, Bergstrom-Pettermann E, Tolmachev V, Hedberg E, Cheng A, Langstrom B. In vitro and animal validation of bromine-76-bromodeoxyuridine as a proliferation marker. J Nucl Med. 1998;39:1273–9. [PubMed] [Google Scholar]
- 58.Loc’h C, Mardon K, Valette H, Brutesco C, Merlet P, Syrota A, Maziere B. Preparation and pharmacological characterization of [76Br]-meta-bromobenzylguanidine ([76Br]MBBG) Nucl Med Biol. 1994;21:49–55. doi: 10.1016/0969-8051(94)90128-7. [DOI] [PubMed] [Google Scholar]
- 59.Scott-Robson S, Capala J, Malmborg P, Lundqvist H. Production of 76Br and its use in labeling proteins. Acta Radiol Suppl. 1991;376:64. [PubMed] [Google Scholar]
- 60.Knight LC, Harwig SL, Welch MJ. In vitro stability and in vivo clearance of fibrinogen or serum albumin labeled with 77Br, 131I, or 125I by direct or indirect synthetic methods. J Nucl Med. 1977;18:282–8. [PubMed] [Google Scholar]
- 61.Wilbur DS, Hylarides MD. Radiolabeling of a monoclonal antibody with N-succinimidyl para-[77Br]bromobenzoate. Int J Rad Appl Instrum B. 1991;18:363–5. doi: 10.1016/0883-2897(91)90133-6. [DOI] [PubMed] [Google Scholar]
- 62.Lovqvist A, Sundin A, Ahlstrom H, Carlsson J, Lundqvist H. 76Br-labeled monoclonal anti-CEA antibodies for radioimmuno positron emission tomography. Nucl Med Biol. 1995;22:125–31. doi: 10.1016/0969-8051(94)e0065-q. [DOI] [PubMed] [Google Scholar]
- 63.Winberg KJ, Persson M, Malmstrom PU, Sjoberg S, Tolmachev V. Radiobromination of anti-HER2/neu/ErbB-2 monoclonal antibody using the p-isothiocyanatobenzene derivative of the [76Br]undecahydro-bromo-7,8-dicarba-nido-undecaborate(1−) ion. Nucl Med Biol. 2004;31:425–33. doi: 10.1016/j.nucmedbio.2003.11.007. [DOI] [PubMed] [Google Scholar]
- 64.Mume E, Orlova A, Larsson B, Nilsson AS, Nilsson FY, Sjoberg S, Tolmachev V. Evaluation of ((4-hydroxyphenyl)ethyl)maleimide for site-specific radiobromination of anti-HER2 affibody. Bioconjug Chem. 2005;16:1547–55. doi: 10.1021/bc050056o. [DOI] [PubMed] [Google Scholar]
- 65.Lovqvist A, Sundin A, Ahlstrom H, Carlsson J, Lundqvist H. Pharmacokinetics and experimental PET imaging of a bromine-76-labeled monoclonal anti-CEA antibody. J Nucl Med. 1997;38:395–401. [PubMed] [Google Scholar]
- 66.Lovqvist A, Lundqvist H, Lubberink M, Tolmachev V, Carlsson J, Sundin A. Kinetics of 76Br-labeled anti-CEA antibodies in pigs; aspects of dosimetry and PET imaging properties. Med Phys. 1999;26:249–58. doi: 10.1118/1.598512. [DOI] [PubMed] [Google Scholar]
- 67.Lovqvist A, Sundin A, Roberto A, Ahlstrom H, Carlsson J, Lundqvist H. Comparative PET imaging of experimental tumors with bromine-76-labeled antibodies, fluorine-18-fluorodeoxyglucose and carbon-11-methionine. J Nucl Med. 1997;38:1029–35. [PubMed] [Google Scholar]
- 68.Hoglund J, Tolmachev V, Orlova A, Lundqvist H, Sundin A. Optimized indirect (76)Br-bromination of antibodies using N-succinimidyl para-[76Br]bromobenzoate for radioimmuno PET. Nucl Med Biol. 2000;27:837–43. doi: 10.1016/s0969-8051(00)00153-0. [DOI] [PubMed] [Google Scholar]
- 69.Maziere B, Loc’h C, Baron JC, Sgouropoulos P, Duquesnoy N, D’Antona R, Cambon H. In vivo quantitative imaging of dopamine receptors in human brain using positron emission tomography and [76Br]bromospiperone. Eur J Pharmacol. 1985;114:267–72. doi: 10.1016/0014-2999(85)90369-3. [DOI] [PubMed] [Google Scholar]
- 70.Martinot JL, Peron-Magnan P, Huret JD, Mazoyer B, Baron JC, Boulenger JP, Loc’h C, Maziere B, Caillard V, Loo H, et al. Striatal D2 dopaminergic receptors assessed with positron emission tomography and [76Br]bromospiperone in untreated schizophrenic patients. Am J Psychiatry. 1990;147:44–50. doi: 10.1176/ajp.147.1.44. [DOI] [PubMed] [Google Scholar]
- 71.Davidsson L, Cederblad A, Hagebo E, Lonnerdal B, Sandstrom B. Intrinsic and extrinsic labeling for studies of manganese absorption in humans. J Nutr. 1988;118:1517–21. doi: 10.1093/jn/118.12.1517. [DOI] [PubMed] [Google Scholar]
- 72.Sastri CS, Petri H, Kueppers G, Erdtmann G. Production of manganese-52 of high isotopic purity by 3He-activation of vanadium. Int J Applied Radiation and Isotopes. 1981;32:246–247. [Google Scholar]
- 73.Lambrecht RM, Wolf AP. The cyclotron production of 38K, 51Mn, 52Mn, and 77Kr for positron emission tomography. Journal of Labelled Compounds and Radiopharmaceuticals. 1979;16:129–130. [Google Scholar]
- 74.Kong G, Jackson C, Koh DM, Lewington V, Sharma B, Brown G, Cunningham D, Cook GJR. The use of 18F-FDG PET/CT in colorectal liver metastases-comparison with CT and liver MRI. European Journal of Nuclear Medicine and Molecular Imaging. 2008:1–7. doi: 10.1007/s00259-008-0743-z. [DOI] [PubMed] [Google Scholar]
- 75.Cikrt M. Biliary excretion of 203 Hg, 64 Cu, 52 Mn, and 210 Pb in the rat. Br J Ind Med. 1972;29:74–80. [PMC free article] [PubMed] [Google Scholar]
- 76.Sharma HZJ, Smith AM, Downey S. Production of cobalt-55, a short-lived, positron emitting radiolabel for bleomycin. International Journal of Radiation and Instrumentation [A] 1986;37:105–109. doi: 10.1016/0883-2889(86)90055-9. [DOI] [PubMed] [Google Scholar]
- 77.Srivastava SCMLF, Kolsky KL, Mease RC, Joshi V, Meinken GE, Pyatt B, Wolf AP, Schlyder DJ, Levy AV, Fowler JS. Production and use of Cobalt-55 as an antibody label for PET imaging. Journal of Labelled Compounds and Radiopharmaceuticals. 1994;35:389–391. [Google Scholar]
- 78.Lagunas-Solar MC, Jungerman JA. Cyclotron production of carrier-free cobalt-55, a new positron-emitting label for bleomycin. International Journal of Applied Radiation and Isotopes. 1979;30:25–32. [Google Scholar]
- 79.Goethals P, Volkaert A, Vandewielle C, Dierckx R, Lameire N. 55Co-EDTA for renal imaging using positron emission tomography (PET): a feasibility study. Nucl Med Biol. 2000;27:77–81. doi: 10.1016/s0969-8051(99)00077-3. [DOI] [PubMed] [Google Scholar]
- 80.Nieweg OE, Beekhuis H, Paans AM, Piers DA, Vaalburg W, Welleweerd J, Wiegman T, Woldring MG. Detection of lung cancer with 55Co-bleomycin using a positron camera. A comparison with 57Co-bleomycin and 55Co-bleomycin single photon scintigraphy. Eur J Nucl Med. 1982;7:104–7. doi: 10.1007/BF00256396. [DOI] [PubMed] [Google Scholar]
- 81.Stevens H, Jansen HM, De Reuck J, Lemmerling M, Strijckmans K, Goethals P, Lemahieu I, de Jong BM, Willemsen AT, Korf J. 55Cobalt (Co) as a PET-tracer in stroke, compared with blood flow, oxygen metabolism, blood volume and gadolinium-MRI. J Neurol Sci. 1999;171:11–8. doi: 10.1016/s0022-510x(99)00229-4. [DOI] [PubMed] [Google Scholar]
- 82.Jansen HM, Paans AM, vd Vliet AM, Veenma-van der Duin L, Bolwijn-Meijer CJ, Pruim J, Willemsen AT, Franssen EJ, Minderhoud JM, Korf J. Cobalt-55 positron emission tomography in ischemic stroke. Clin Neurol Neurosurg. 1997;99:6–10. doi: 10.1016/s0303-8467(96)00558-6. [DOI] [PubMed] [Google Scholar]
- 83.Byegård JSG, Skålberg M. The stability of some metal EDTA, DTPA and DOTA complexes: Application as tracers in groundwater studies. Journal of Radioanalytical and Nuclear Chemistry. 1999;241:281–290. [Google Scholar]
- 84.Heppeler A, Andre JP, Buschmann I, Wang X, Reubi JC, Hennig M, Kaden TA, Maecke HR. Metal-ion-dependent biological properties of a chelator-derived somatostatin analogue for tumour targeting. Chemistry. 2008;14:3026–34. doi: 10.1002/chem.200701264. [DOI] [PubMed] [Google Scholar]
- 85.Kukis DL, DeNardo SJ, DeNardo GL, O’Donnell RT, Meares CF. Optimized conditions for chelation of yttrium-90-DOTA immunoconjugates. J Nucl Med. 1998;39:2105–10. [PubMed] [Google Scholar]
- 86.Kukis DL, DeNardo GL, DeNardo SJ, Mirick GR, Miers LA, Greiner DP, Meares CF. Effect of the extent of chelate substitution on the immunoreactivity and biodistribution of 2IT-BAT-Lym-1 immunoconjugates. Cancer Res. 1995;55:878–84. [PubMed] [Google Scholar]
- 87.Beekhuis H, Nieweg OE. Radiation absorbed doses from Co-57- and Co-55 bleomycin. J Nucl Med. 1984;25:478–85. [PubMed] [Google Scholar]
- 88.Bowen H. Trace Elements in Biochemistry. Academic Press; London: 1966. The Biogeochemistry of the Elements; pp. 173–239. [Google Scholar]
- 89.Goethals P, Coene M, Slegers G, Vogelaers D, Everaert J, Lemahieu I, Colardyn F, Heyndrickx GR. Production of carrier-free 66Ga and labeling of antimyosin antibody for positron imaging of acute myocardial infarction. Eur J Nucl Med. 1990;16:237–40. doi: 10.1007/BF00842774. [DOI] [PubMed] [Google Scholar]
- 90.Graham MC, Pentlow KS, Mawlawi O, Finn RD, Daghighian F, Larson SM. An investigation of the physical characteristics of 66Ga as an isotope for PET imaging and quantification. Med Phys. 1997;24:317–26. doi: 10.1118/1.597924. [DOI] [PubMed] [Google Scholar]
- 91.Szelecsenyi F, Tarkanyi F, Kovacs Z, Bergman J, Heselius SJ, Solin O. Production of 66Ga and 67Ga at a compact cyclotron. Acta Radiol Suppl. 1991;376:62–3. [PubMed] [Google Scholar]
- 92.Goethals P, Coene M, Slegers G, Agon P, Deman J, Schelstraete K. Cyclotron production of carrier-free 66Ga as a positron emitting label of albumin colloids for clinical use. Eur J Nucl Med. 1988;14:152–4. doi: 10.1007/BF00293540. [DOI] [PubMed] [Google Scholar]
- 93.Zweit J, Sharma H, Downey S. Production of gallium-66, a short-lived, positron emitting radionuclide. Int J Rad Appl Instrum [A] 1987;38:499–501. doi: 10.1016/0883-2889(87)90194-8. [DOI] [PubMed] [Google Scholar]
- 94.Green MA, Welch MJ. Gallium radiopharmaceutical chemistry. Int J Rad Appl Instrum B. 1989;16:435–48. doi: 10.1016/0883-2897(89)90053-6. [DOI] [PubMed] [Google Scholar]
- 95.Brechbiel MW. Bifunctional chelates for metal nuclides. Q J Nucl Med Mol Imaging. 2008;52:166–73. [PMC free article] [PubMed] [Google Scholar]
- 96.Harris WR, Chen Y. Electron paramagnetic resonance and difference ultraviolet studies of Mn2+ binding to serum transferrin. J Inorg Biochem. 1994;54:1–19. doi: 10.1016/0162-0134(94)85119-0. [DOI] [PubMed] [Google Scholar]
- 97.Eisenwiener KP, Prata MI, Buschmann I, Zhang HW, Santos AC, Wenger S, Reubi JC, Macke HR. NODAGATOC, a new chelator-coupled somatostatin analogue labeled with [67/68Ga] and [111In] for SPECT, PET, and targeted therapeutic applications of somatostatin receptor (hsst2) expressing tumors. Bioconjug Chem. 2002;13:530–41. doi: 10.1021/bc010074f. [DOI] [PubMed] [Google Scholar]
- 98.Lee J, Garmestani K, Wu C, Brechbiel MW, Chang HK, Choi CW, Gansow OA, Carrasquillo JA, Paik CH. In vitro and in vivo evaluation of structure-stability relationship of 111In- and 67Ga-labeled antibody via 1B4M or C-NOTA chelates. Nucl Med Biol. 1997;24:225–30. doi: 10.1016/s0969-8051(97)00056-5. [DOI] [PubMed] [Google Scholar]
- 99.Velikyan I, Maecke H, Langstrom B. Convenient preparation of 68Ga-based PET-radiopharmaceuticals at room temperature. Bioconjug Chem. 2008;19:569–73. doi: 10.1021/bc700341x. [DOI] [PubMed] [Google Scholar]
- 100.Mathias CJ, Lewis MR, Reichert DE, Laforest R, Sharp TL, Lewis JS, Yang ZF, Waters DJ, Snyder PW, Low PS, Welch MJ, Green MA. Preparation of 66Ga- and 68Ga-labeled Ga(III)-deferoxamine-folate as potential folate-receptor-targeted PET radiopharmaceuticals. Nucl Med Biol. 2003;30:725–31. doi: 10.1016/s0969-8051(03)00080-5. [DOI] [PubMed] [Google Scholar]
- 101.Ugur O, Kothari PJ, Finn RD, Zanzonico P, Ruan S, Guenther I, Maecke HR, Larson SM. Ga-66 labeled somatostatin analogue DOTA-DPhe1-Tyr3-octreotide as a potential agent for positron emission tomography imaging and receptor mediated internal radiotherapy of somatostatin receptor positive tumors. Nucl Med Biol. 2002;29:147–57. doi: 10.1016/s0969-8051(01)00290-6. [DOI] [PubMed] [Google Scholar]
- 102.Van Der Werff JT. Clinical investigations on the use of radioactive gallium (Ga66 and Ga67) in bone diseases. Acta radiol. 1954;41:343–7. [PubMed] [Google Scholar]
- 103.Henze M, Dimitrakopoulou-Strauss A, Milker-Zabel S, Schuhmacher J, Strauss LG, Doll J, Macke HR, Eisenhut M, Debus J, Haberkorn U. Characterization of 68Ga-DOTA-D-Phe1-Tyr3-octreotide kinetics in patients with meningiomas. J Nucl Med. 2005;46:763–9. [PubMed] [Google Scholar]
- 104.Koukouraki S, Strauss LG, Georgoulias V, Eisenhut M, Haberkorn U, Dimitrakopoulou-Strauss A. Comparison of the pharmacokinetics of 68Ga-DOTATOC and [18F]FDG in patients with metastatic neuroendocrine tumours scheduled for 90Y-DOTATOC therapy. Eur J Nucl Med Mol Imaging. 2006;33:1115–22. doi: 10.1007/s00259-006-0110-x. [DOI] [PubMed] [Google Scholar]
- 105.Smith-Jones PM, Solit D, Afroze F, Rosen N, Larson SM. Early tumor response to Hsp90 therapy using HER2 PET: comparison with 18F-FDG PET. J Nucl Med. 2006;47:793–6. [PMC free article] [PubMed] [Google Scholar]
- 106.Smith-Jones PM, Solit DB, Akhurst T, Afroze F, Rosen N, Larson SM. Imaging the pharmacodynamics of HER2 degradation in response to Hsp90 inhibitors. Nat Biotechnol. 2004;22:701–6. doi: 10.1038/nbt968. [DOI] [PubMed] [Google Scholar]
- 107.Jennewein M, Qaim SM, Hermanne A, Jahn M, Tsyganov E, Slavine N, Seliounine S, Antich PA, Kulkarni PV, Thorpe PE, Mason RP, Rösch F. A new method for radiochemical separation of arsenic from irradiated germanium oxide. Applied Radiation and Isotopes. 2005;63:343–351. doi: 10.1016/j.apradiso.2005.04.005. [DOI] [PubMed] [Google Scholar]
- 108.Jennewein M, Qaim SM, Kulkarni PV, Mason RP, Hermanne A, Rösch F. A no-carrier-added 72Se/72As radionuclide generator based on solid phase extraction. Radiochimica Acta. 2005;93:579–583. [Google Scholar]
- 109.Jennewein M, Schmidt A, Novgorodov AF, Qaim SM, Rösch F. A no-carrier-added 72Se/72As radionuclide generator based on distillation. Radiochimica Acta. 2004;92:245–249. [Google Scholar]
- 110.Al-Kouraishi SH, Boswell GGJ. An isotope generator for 72As. International Journal of Applied Radiation and Isotopes. 1978;29:607–609. [Google Scholar]
- 111.Jennewein M, Lewis MA, Zhao D, Tsyganov E, Slavine N, He J, Watkins L, Kodibagkar VD, O’Kelly S, Kulkarni P, Antich PP, Hermanne A, Rosch F, Mason RP, Thorpe PE. Vascular Imaging of Solid Tumors in Rats with a Radioactive Arsenic-Labeled Antibody that Binds Exposed Phosphatidylserine. Clin Cancer Res. 2008;14:1377–85. doi: 10.1158/1078-0432.CCR-07-1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Turner PC. The calculation of absorbed tissue doses from arsenic-72. Phys Med Biol. 1962;7:329–34. doi: 10.1088/0031-9155/7/3/305. [DOI] [PubMed] [Google Scholar]
- 113.Connett JM, Buettner TL, Anderson CJ. Maximum tolerated dose and large tumor radioimmunotherapy studies of 64Cu-labeled monoclonal antibody 1A3 in a colon cancer model. Clin Cancer Res. 1999;5:3207s–3212s. [PubMed] [Google Scholar]
- 114.Ruangma A, Bai B, Lewis JS, Sun X, Welch MJ, Leahy R, Laforest R. Three-dimensional maximum a posteriori (MAP) imaging with radiopharmaceuticals labeled with three Cu radionuclides. Nucl Med Biol. 2006;33:217–26. doi: 10.1016/j.nucmedbio.2005.11.001. [DOI] [PubMed] [Google Scholar]
- 115.Obata A, Kasamatsu S, McCarthy DW, Welch MJ, Saji H, Yonekura Y, Fujibayashi Y. Production of therapeutic quantities of (64)Cu using a 12 MeV cyclotron. Nucl Med Biol. 2003;30:535–9. doi: 10.1016/s0969-8051(03)00024-6. [DOI] [PubMed] [Google Scholar]
- 116.McCarthy DW, Shefer RE, Klinkowstein RE, Bass LA, Margeneau WH, Cutler CS, Anderson CJ, Welch MJ. Efficient production of high specific activity 64Cu using a biomedical cyclotron. Nucl Med Biol. 1997;24:35–43. doi: 10.1016/s0969-8051(96)00157-6. [DOI] [PubMed] [Google Scholar]
- 117.Zinn KR, Chaudhuri TR, Cheng TP, Morris JS, Meyer WA., Jr Production of no-carrier-added 64Cu from zinc metal irradiated under boron shielding. Cancer. 1994;73:774–8. doi: 10.1002/1097-0142(19940201)73:3+<774::aid-cncr2820731305>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 118.Hilgers K, Stoll T, Skakun Y, Coenen HH, Qaim SM. Cross-section measurements of the nuclear reactions natZn(d,x)64Cu, 66Zn(d,alpha)64Cu and 68Zn(p,alphan)64Cu for production of 64Cu and technical developments for small-scale production of 67Cu via the 70Zn(p,alpha)67Cu process. Appl Radiat Isot. 2003;59:343–51. doi: 10.1016/s0969-8043(03)00199-4. [DOI] [PubMed] [Google Scholar]
- 119.Qaim SM. Decay data and production yields of some non-standard positron emitters used in PET. Q J Nucl Med Mol Imaging. 2008;52:111–6. [PubMed] [Google Scholar]
- 120.Szelecsényi F, Steyn GF, Kovács Z, Vermeulen C, Van Der Meulen NP, Dolley SG, Van Der Walt TN, Suzuki K, Mukai K. Investigation of the 66Zn(p,2pn)64Cu and 68Zn(p,x)64Cu nuclear processes up to 100 MeV: Production of 64Cu. Nuclear Instruments and Methods in Physics Research, Section B: Beam Interactions with Materials and Atoms. 2005;240:625–637. [Google Scholar]
- 121.Cole WC, DeNardo SJ, Meares CF, McCall MJ, DeNardo GL, Epstein AL, O’Brien HA, Moi MK. Serum stability of 67Cu chelates: comparison with 111In and 57Co. Int J Rad Appl Instrum B. 1986;13:363–8. doi: 10.1016/0883-2897(86)90011-5. [DOI] [PubMed] [Google Scholar]
- 122.Franz J, Freeman GM, Barefield EK, Volkert WA, Ehrhardt GJ, Holmes RA. Labeling of antibodies with 64Cu using a conjugate containing a macrocyclic amine chelating agent. International Journal of Radiation Applications and Instrumentation. Part B Nuclear Medicine and Biology. 1987;14:479–484. doi: 10.1016/0883-2897(87)90113-9. [DOI] [PubMed] [Google Scholar]
- 123.Rogers BE, Anderson CJ, Connett JM, Guo LW, Edwards WB, Sherman EL, Zinn KR, Welch MJ. Comparison of four bifunctional chelates for radiolabeling monoclonal antibodies with copper radioisotopes: biodistribution and metabolism. Bioconjug Chem. 1996;7:511–22. doi: 10.1021/bc9600372. [DOI] [PubMed] [Google Scholar]
- 124.Anderson CJ, Connett JM, Schwarz SW, Rocque PA, Guo LW, Philpott GW, Zinn KR, Meares CF, Welch MJ. Copper-64-labeled antibodies for PET imaging. J Nucl Med. 1992;33:1685–91. [PubMed] [Google Scholar]
- 125.Ping Li W, Meyer LA, Capretto DA, Sherman CD, Anderson CJ. Receptor-binding, biodistribution, and metabolism studies of 64Cu-DOTA-cetuximab, a PET-imaging agent for epidermal growth-factor receptor-positive tumors. Cancer Biother Radiopharm. 2008;23:158–71. doi: 10.1089/cbr.2007.0444. [DOI] [PubMed] [Google Scholar]
- 126.Anderson CJ, Wadas TJ, Wong EH, Weisman GR. Cross-bridged macrocyclic chelators for stable complexation of copper radionuclides for PET imaging. Q J Nucl Med Mol Imaging. 2008;52:185–92. [PMC free article] [PubMed] [Google Scholar]
- 127.Sprague JE, Peng Y, Sun X, Weisman GR, Wong EH, Achilefu S, Anderson CJ. Preparation and biological evaluation of copper-64-labeled tyr3-octreotate using a cross-bridged macrocyclic chelator. Clin Cancer Res. 2004;10:8674–82. doi: 10.1158/1078-0432.CCR-04-1084. [DOI] [PubMed] [Google Scholar]
- 128.Voss SD, Smith SV, DiBartolo N, McIntosh LJ, Cyr EM, Bonab AA, Dearling JL, Carter EA, Fischman AJ, Treves ST, Gillies SD, Sargeson AM, Huston JS, Packard AB. Positron emission tomography (PET) imaging of neuroblastoma and melanoma with 64Cu-SarAr immunoconjugates. Proc Natl Acad Sci U S A. 2007;104:17489–93. doi: 10.1073/pnas.0708436104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Bhalgat MK, Roberts JC, Mercer-Smith JA, Knotts BD, Vessella RL, Lavallee DK. Preparation and biodistribution of copper-67-labeled porphyrins and porphyrin-A6H immunoconjugates. Nucl Med Biol. 1997;24:179–85. doi: 10.1016/s0969-8051(96)00215-6. [DOI] [PubMed] [Google Scholar]
- 130.Ping Li W, Meyer LA, Capretto DA, Sherman CD, Anderson CJ. Receptor-Binding, Biodistribution, and Metabolism Studies of (64)Cu-DOTA-Cetuximab, a PET-Imaging Agent for Epidermal Growth-Factor Receptor-Positive Tumors. Cancer Biother Radiopharm. 2008;23:158–71. doi: 10.1089/cbr.2007.0444. [DOI] [PubMed] [Google Scholar]
- 131.Cai W, Wu Y, Chen K, Cao Q, Tice DA, Chen X. In vitro and in vivo characterization of 64Cu-labeled Abegrin, a humanized monoclonal antibody against integrin alpha v beta 3. Cancer Res. 2006;66:9673–81. doi: 10.1158/0008-5472.CAN-06-1480. [DOI] [PubMed] [Google Scholar]
- 132.Cai W, Chen K, He L, Cao Q, Koong A, Chen X. Quantitative PET of EGFR expression in xenograft-bearing mice using 64Cu-labeled cetuximab, a chimeric anti-EGFR monoclonal antibody. Eur J Nucl Med Mol Imaging. 2007;34:850–8. doi: 10.1007/s00259-006-0361-6. [DOI] [PubMed] [Google Scholar]
- 133.Grunberg J, Novak-Hofer I, Honer M, Zimmermann K, Knogler K, Blauenstein P, Ametamey S, Maecke HR, Schubiger PA. In vivo evaluation of 177Lu- and 67/64Cu-labeled recombinant fragments of antibody chCE7 for radioimmunotherapy and PET imaging of L1-CAM-positive tumors. Clin Cancer Res. 2005;11:5112–20. doi: 10.1158/1078-0432.CCR-05-0227. [DOI] [PubMed] [Google Scholar]
- 134.Novak-Hofer I, Honer M, Ametamey S, Grunberg J, Missimer J, Zimmermann K, Schubiger PA. Imaging of renal carcinoma xenografts with 64Cu-labelled anti-L1-CAM antibody chCE7. Eur J Nucl Med Mol Imaging. 2003;30:1066. doi: 10.1007/s00259-003-1222-1. [DOI] [PubMed] [Google Scholar]
- 135.Li L, Bading J, Yazaki PJ, Ahuja AH, Crow D, Colcher D, Williams LE, Wong JY, Raubitschek A, Shively JE. A versatile bifunctional chelate for radiolabeling humanized anti-CEA antibody with In-111 and Cu-64 at either thiol or amino groups: PET imaging of CEA-positive tumors with whole antibodies. Bioconjug Chem. 2008;19:89–96. doi: 10.1021/bc700161p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Connett JM, Anderson CJ, Guo LW, Schwarz SW, Zinn KR, Rogers BE, Siegel BA, Philpott GW, Welch MJ. Radioimmunotherapy with a 64Cu-labeled monoclonal antibody: a comparison with 67Cu. Proc Natl Acad Sci U S A. 1996;93:6814–8. doi: 10.1073/pnas.93.13.6814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Philpott GW, Schwarz SW, Anderson CJ, Dehdashti F, Connett JM, Zinn KR, Meares CF, Cutler PD, Welch MJ, Siegel BA. RadioimmunoPET: detection of colorectal carcinoma with positron-emitting copper-64-labeled monoclonal antibody. J Nucl Med. 1995;36:1818–24. [PubMed] [Google Scholar]
- 138.Yoo J, Tang L, Perkins TA, Rowland DJ, Laforest R, Lewis JS, Welch MJ. Preparation of high specific activity (86)Y using a small biomedical cyclotron. Nucl Med Biol. 2005;32:891–7. doi: 10.1016/j.nucmedbio.2005.06.007. [DOI] [PubMed] [Google Scholar]
- 139.Garmestani K, Milenic DE, Plascjak PS, Brechbiel MW. A new and convenient method for purification of 86Y using a Sr(II) selective resin and comparison of biodistribution of 86Y and 111In labeled Herceptin. Nucl Med Biol. 2002;29:599–606. doi: 10.1016/s0969-8051(02)00322-0. [DOI] [PubMed] [Google Scholar]
- 140.Park LS, Szajek LP, Wong KJ, Plascjak PS, Garmestani K, Googins S, Eckelman WC, Carrasquillo JA, Paik CH. Semi-automated 86Y purification using a three-column system. Nucl Med Biol. 2004;31:297–301. doi: 10.1016/j.nucmedbio.2003.07.002. [DOI] [PubMed] [Google Scholar]
- 141.Smith S, Thomas RM, Steere HA, Beatty HE, Dawson KB, Peckham MJ. Therapeutic irradiation of the central nervous system using intrathecal 90Y--DTA. Br J Radiol. 1976;49:141–7. doi: 10.1259/0007-1285-49-578-141. [DOI] [PubMed] [Google Scholar]
- 142.Hnatowich DJ, Virzi F, Doherty PW. DTPA-coupled antibodies labeled with yttrium-90. J Nucl Med. 1985;26:503–9. [PubMed] [Google Scholar]
- 143.Sharkey RM, Motta-Hennessy C, Gansow OA, Brechbiel MW, Fand I, Griffiths GL, Jones AL, Goldenberg DM. Selection of a DTPA chelate conjugate for monoclonal antibody targeting to a human colonic tumor in nude mice. Int J Cancer. 1990;46:79–85. doi: 10.1002/ijc.2910460116. [DOI] [PubMed] [Google Scholar]
- 144.Washburn LC, Sun TT, Lee YC, Byrd BL, Holloway EC, Crook JE, Stubbs JB, Stabin MG, Brechbiel MW, Gansow OA, et al. Comparison of five bifunctional chelate techniques for 90Y-labeled monoclonal antibody CO17–1A. Int J Rad Appl Instrum B. 1991;18:313–21. doi: 10.1016/0883-2897(91)90127-7. [DOI] [PubMed] [Google Scholar]
- 145.Brechbiel MW, Gansow OA. Backbone-substituted DTPA ligands for 90Y radioimmunotherapy. Bioconjug Chem. 1991;2:187–94. doi: 10.1021/bc00009a008. [DOI] [PubMed] [Google Scholar]
- 146.Camera L, Kinuya S, Garmestani K, Wu C, Brechbiel MW, Pai LH, McMurry TJ, Gansow OA, Pastan I, Paik CH, et al. Evaluation of the serum stability and in vivo biodistribution of CHX-DTPA and other ligands for yttrium labeling of monoclonal antibodies. J Nucl Med. 1994;35:882–9. [PubMed] [Google Scholar]
- 147.Camera L, Kinuya S, Garmestani K, Pai LH, Brechbiel MW, Gansow OA, Paik CH, Pastan I, Carrasquillo JA. Evaluation of a new DTPA-derivative chelator: comparative biodistribution and imaging studies of 111In-labeled B3 monoclonal antibody in athymic mice bearing human epidermoid carcinoma xenografts. Nucl Med Biol. 1993;20:955–62. doi: 10.1016/0969-8051(93)90096-d. [DOI] [PubMed] [Google Scholar]
- 148.Kobayashi H, Wu C, Yoo TM, Sun BF, Drumm D, Pastan I, Paik CH, Gansow OA, Carrasquillo JA, Brechbiel MW. Evaluation of the in vivo biodistribution of yttrium-labeled isomers of CHX-DTPA-conjugated monoclonal antibodies. J Nucl Med. 1998;39:829–36. [PubMed] [Google Scholar]
- 149.Deshpande SV, DeNardo SJ, Kukis DL, Moi MK, McCall MJ, DeNardo GL, Meares CF. Yttrium-90-labeled monoclonal antibody for therapy: labeling by a new macrocyclic bifunctional chelating agent. J Nucl Med. 1990;31:473–9. [PubMed] [Google Scholar]
- 150.Harrison A, Walker CA, Parker D, Jankowski KJ, Cox JP, Craig AS, Sansom JM, Beeley NR, Boyce RA, Chaplin L, et al. The in vivo release of 90Y from cyclic and acyclic ligand-antibody conjugates. Int J Rad Appl Instrum B. 1991;18:469–76. doi: 10.1016/0883-2897(91)90107-v. [DOI] [PubMed] [Google Scholar]
- 151.Wong JY, Chu DZ, Williams LE, Liu A, Zhan J, Yamauchi DM, Wilczynski S, Wu AM, Yazaki PJ, Shively JE, Leong L, Raubitschek AA. A phase I trial of (90)Y-DOTA-anti-CEA chimeric T84.66 (cT84.66) radioimmunotherapy in patients with metastatic CEA-producing malignancies. Cancer Biother Radiopharm. 2006;21:88–100. doi: 10.1089/cbr.2006.21.88. [DOI] [PubMed] [Google Scholar]
- 152.Palm S, Enmon RM, Jr, Matei C, Kolbert KS, Xu S, Zanzonico PB, Finn RL, Koutcher JA, Larson SM, Sgouros G. Pharmacokinetics and Biodistribution of (86)Y-Trastuzumab for (90)Y dosimetry in an ovarian carcinoma model: correlative MicroPET and MRI. J Nucl Med. 2003;44:1148–55. [PubMed] [Google Scholar]
- 153.Lovqvist A, Humm JL, Sheikh A, Finn RD, Koziorowski J, Ruan S, Pentlow KS, Jungbluth A, Welt S, Lee FT, Brechbiel MW, Larson SM. PET imaging of (86)Y-labeled anti-Lewis Y monoclonal antibodies in a nude mouse model: comparison between (86)Y and (111)In radiolabels. J Nucl Med. 2001;42:1281–7. [PubMed] [Google Scholar]
- 154.Regino CM, Wong D, Garmestani K, Baidoo K, Williams K, Seidel M, Green J, Choyke M, Brechbiel PM. 99th Annual Meeting of the American Association for Cancer Research. American Association for Cancer Research; San Diego: 2008. p. Abstract # 3159. [Google Scholar]
- 155.Jamar F, Barone R, Mathieu I, Walrand S, Labar D, Carlier P, de Camps J, Schran H, Chen T, Smith MC, Bouterfa H, Valkema R, Krenning EP, Kvols LK, Pauwels S. 86Y-DOTA0)-D-Phe1-Tyr3-octreotide (SMT487)--a phase 1 clinical study: pharmacokinetics, biodistribution and renal protective effect of different regimens of amino acid co-infusion. Eur J Nucl Med Mol Imaging. 2003;30:510–8. doi: 10.1007/s00259-003-1117-1. [DOI] [PubMed] [Google Scholar]
- 156.Perk LR, Visser GW, Vosjan MJ, Stigter-van Walsum M, Tijink BM, Leemans CR, van Dongen GA. (89)Zr as a PET surrogate radioisotope for scouting biodistribution of the therapeutic radiometals (90)Y and (177)Lu in tumor-bearing nude mice after coupling to the internalizing antibody cetuximab. J Nucl Med. 2005;46:1898–906. [PubMed] [Google Scholar]
- 157.Hohn A, Zimmermann K, Schaub E, Hirzel W, Schubiger PA, Schibli R. Production and separation of “non-standard” positron emission tomography nuclides at a large cyclotron facility: the experiences at the Paul Scherrer Institute Switzerland. Q J Nucl Med Mol Imaging. 2008 [PubMed] [Google Scholar]
- 158.Meijs WE, Herscheid JD, Haisma HJ, Pinedo HM. Evaluation of desferal as a bifunctional chelating agent for labeling antibodies with Zr-89. Int J Rad Appl Instrum [A] 1992;43:1443–7. doi: 10.1016/0883-2889(92)90170-j. [DOI] [PubMed] [Google Scholar]
- 159.Pagani M, Stone-Elander S, Larsson SA. Alternative positron emission tomography with non-conventional positron emitters: effects of their physical properties on image quality and potential clinical applications. Eur J Nucl Med. 1997;24:1301–27. doi: 10.1007/s002590050156. [DOI] [PubMed] [Google Scholar]
- 160.Link JM, Krohn KA, Eary JF. 89Zr for antibody labeling and positron emission tomography. Journal of Labelled Compounds and Radiopharmaceuticals. 1986;23:1297–1298. [Google Scholar]
- 161.Ali SA, Shankland EG. Radiochemical synthesis of 89Zr-porphyrin for positron label of monoclonal antibodies. Journal of Labelled Compounds and Radiopharmaceuticals. 1991;30:326. [Google Scholar]
- 162.Meijs WE, Haisma HJ, Klok RP, van Gog FB, Kievit E, Pinedo HM, Herscheid JD. Zirconium-labeled monoclonal antibodies and their distribution in tumor-bearing nude mice. J Nucl Med. 1997;38:112–8. [PubMed] [Google Scholar]
- 163.Meijs WE, Haisma HJ, Van der Schors R, Wijbrandts R, Van den Oever K, Klok RP, Pinedo HM, Herscheid JD. A facile method for the labeling of proteins with zirconium isotopes. Nucl Med Biol. 1996;23:439–48. doi: 10.1016/0969-8051(96)00020-0. [DOI] [PubMed] [Google Scholar]
- 164.Verel I, Visser GW, Boellaard R, Stigter-van Walsum M, Snow GB, van Dongen GA. 89Zr immuno-PET: comprehensive procedures for the production of 89Zr-labeled monoclonal antibodies. J Nucl Med. 2003;44:1271–81. [PubMed] [Google Scholar]
- 165.Witzig TE, Gordon LI, Cabanillas F, Czuczman MS, Emmanouilides C, Joyce R, Pohlman BL, Bartlett NL, Wiseman GA, Padre N, Grillo-Lopez AJ, Multani P, White CA. Randomized controlled trial of yttrium-90-labeled ibritumomab tiuxetan radioimmunotherapy versus rituximab immunotherapy for patients with relapsed or refractory low-grade, follicular, or transformed B-cell non-Hodgkin’s lymphoma. J Clin Oncol. 2002;20:2453–63. doi: 10.1200/JCO.2002.11.076. [DOI] [PubMed] [Google Scholar]
- 166.Verel I, Visser GW, van Dongen GA. The promise of immuno-PET in radioimmunotherapy. J Nucl Med. 2005;46(Suppl 1):164S–71S. [PubMed] [Google Scholar]
- 167.Nagengast WB, de Vries EG, Hospers GA, Mulder NH, de Jong JR, Hollema H, Brouwers AH, van Dongen GA, Perk LR, Lub-de Hooge MN. In vivo VEGF imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft. J Nucl Med. 2007;48:1313–9. doi: 10.2967/jnumed.107.041301. [DOI] [PubMed] [Google Scholar]
- 168.Perk LR, Visser OJ, Stigter-van Walsum M, Vosjan MJ, Visser GW, Zijlstra JM, Huijgens PC, van Dongen GA. Preparation and evaluation of (89)Zr-Zevalin for monitoring of (90)Y-Zevalin biodistribution with positron emission tomography. Eur J Nucl Med Mol Imaging. 2006;33:1337–45. doi: 10.1007/s00259-006-0160-0. [DOI] [PubMed] [Google Scholar]
- 169.Borjesson PK, Jauw YW, Boellaard R, de Bree R, Comans EF, Roos JC, Castelijns JA, Vosjan MJ, Kummer JA, Leemans CR, Lammertsma AA, van Dongen GA. Performance of immuno-positron emission tomography with zirconium-89-labeled chimeric monoclonal antibody U36 in the detection of lymph node metastases in head and neck cancer patients. Clin Cancer Res. 2006;12:2133–40. doi: 10.1158/1078-0432.CCR-05-2137. [DOI] [PubMed] [Google Scholar]
- 170.Perk LR, Stigter-van Walsum M, Visser GW, Kloet RW, Vosjan MJ, Leemans CR, Giaccone G, Albano R, Comoglio PM, van Dongen GA. Quantitative PET imaging of Met-expressing human cancer xenografts with (89)Zr-labelled monoclonal antibody DN30. Eur J Nucl Med Mol Imaging. 2008 doi: 10.1007/s00259-008-0774-5. [DOI] [PubMed] [Google Scholar]
- 171.Laforest R, Liu X. Image quality with non-standard nuclides in positron emission tomography. Q J Nucl Med Mol Imaging. 2007 [PubMed] [Google Scholar]
- 172.Beattie BJ, Finn RD, Rowland DJ, Pentlow KS. Quantitative imaging of bromine-76 and yttrium-86 with PET: a method for the removal of spurious activity introduced by cascade gamma rays. Med Phys. 2003;30:2410–23. doi: 10.1118/1.1595599. [DOI] [PubMed] [Google Scholar]
- 173.Vandenberghe S. Three-dimensional positron emission tomography imaging with 124I and 86Y. Nucl Med Commun. 2006;27:237–45. doi: 10.1097/01.mnm.0000199476.46525.2c. [DOI] [PubMed] [Google Scholar]
- 174.Bale WF, Spar IL. Studies directed toward the use of antibodies as carriers of radioactivity for therapy. Adv Biol Med Phys. 1957;5:285–356. doi: 10.1016/b978-1-4832-3111-2.50011-0. [DOI] [PubMed] [Google Scholar]
- 175.Bale WF, Spar IL, Goodland RL. Experimental radiation therapy of tumors with I-131-carrying antibodies to fibrin. Cancer Res. 1960;20:1488–94. [PubMed] [Google Scholar]
- 176.Milenic DE, Brady ED, Brechbiel MW. Antibody-targeted radiation cancer therapy. Nat Rev Drug Discov. 2004;3:488–99. doi: 10.1038/nrd1413. [DOI] [PubMed] [Google Scholar]
- 177.O’Donoghue JA. Implications of nonuniform tumor doses for radioimmunotherapy. J Nucl Med. 1999;40:1337–41. [PubMed] [Google Scholar]
- 178.Howell RW, Goddu SM, Rao DV. Application of the linear-quadratic model to radioimmunotherapy: further support for the advantage of longer-lived radionuclides. J Nucl Med. 1994;35:1861–9. [PubMed] [Google Scholar]
- 179.Fowler JF. Radiobiological aspects of low dose rates in radioimmunotherapy. Int J Radiat Oncol Biol Phys. 1990;18:1261–9. doi: 10.1016/0360-3016(90)90467-x. [DOI] [PubMed] [Google Scholar]
- 180.Nunn AD. Molecular imaging and personalized medicine: an uncertain future. Cancer Biother Radiopharm. 2007;22:722–39. doi: 10.1089/cbr.2007.0417. [DOI] [PubMed] [Google Scholar]
- 181.Brouwers AH, Dorr U, Lang O, Boerman OC, Oyen WJ, Steffens MG, Oosterwijk E, Mergenthaler HG, Bihl H, Corstens FH. 131 I-cG250 monoclonal antibody immunoscintigraphy versus [18 F]FDG-PET imaging in patients with metastatic renal cell carcinoma: a comparative study. Nucl Med Commun. 2002;23:229–36. doi: 10.1097/00006231-200203000-00005. [DOI] [PubMed] [Google Scholar]
- 182.Rahmim A, Zaidi H. PET versus SPECT: strengths, limitations and challenges. Nucl Med Commun. 2008;29:193–207. doi: 10.1097/MNM.0b013e3282f3a515. [DOI] [PubMed] [Google Scholar]
- 183.Beekman F, van der Have F. The pinhole: gateway to ultra-high-resolution three-dimensional radionuclide imaging. Eur J Nucl Med Mol Imaging. 2007;34:151–61. doi: 10.1007/s00259-006-0248-6. [DOI] [PubMed] [Google Scholar]