Abstract
Objectives
To compare the radiolabeling performance, stability, and practical efficacy of the chelators CHX-A″-DTPA and H4octapa with the therapeutic radiometal 90Y.
Methods
The bifunctional chelators p-SCN-Bn-H4octapa and p-SCN-Bn-CHX-A″-DTPA were conjugated to the HER2-targeting antibody trastuzumab. The resulting immunoconjugates were radiolabeled with 90Y to compare radiolabeling efficiency, in vitro and in vivo stability, and in vivo performance in a murine model of ovarian cancer.
Results
High radiochemical yields (>95%) were obtained with 90Y-CHX-A′-DTPA-trastuzumab and 90Y-octapa-trastuzumab after 15 min at room temperature. Both 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab exhibited excellent in vitro and in vivo stability. Furthermore, the radioimmunoconjugates displayed high tumoral uptake values (42.3 ± 4.0%ID/g for 90Y-CHX-A″-DTPA-trastuzumab and 30.1 ± 7.4%ID/g for 90Y-octapa-trastuzumab at 72 h post-injection) in mice bearing HER2-expressing SKOV3 ovarian cancer xenografts. Finally, 90Y radioimmunotherapy studies performed in tumor-bearing mice demonstrated that 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab are equally effective therapeutic agents, as treatment with both radioimmunoconjugates yielded substantially decreased tumor growth compared to controls.
Conclusions
Ultimately, this work demonstrates that the acyclic chelators CHX-A″-DTPA and H4octapa have comparable radiolabeling, stability, and in vivo performance, making them both suitable choices for applications requiring 90Y.
Keywords: Yttrium-90, Trastuzumab, CHX-A″-DTPA, H4octapa, Radiometal, Radioimmunotherapy
1. Introduction
The β−-emitting radiometal 90Y is commonly used for peptide receptor radionuclide therapy (PRRT, e.g., 90Y-DOTATOC) and radioimmunotherapy (e.g., 90Y-DOTA-trastuzumab) to deliver a therapeutic dose of radiation to tumor cells [1,2]. One advantage of 90Y (t1/2=64.1 h, β−=72%, MeV) compared to other therapeutic isotopes is that it can treat relatively large and poorly vascularized tumors due to its long in vivo β− range of ~12 mm [3–5]. This relatively long particle range means that tumor cells up to ~550 cell diameters away from the radiopharmaceutical will also receive therapeutic irradiation, a phenomenon known as the “crossfire effect” [3–5]. While the crossfire effect is generally regarded as positive due to the ability to treat tumors of larger sizes, an inevitable disadvantage of the long β− range is the potential irradiation of healthy tissue near the tumor.
Critically, the use of 90Y for PRRT and radioimmunotherapy necessitates the use of a pre-therapy scout imaging scan to perform dosimetry, as 90Y itself emits no particles or gamma rays suitable for imaging [6,7]. In this type of scout imaging, 90Y is replaced with an isotope appropriate for imaging: for example, 111In-DOTATOC will be used as an imaging counterpart to 90Y-DOTATOC [6,7]. A number of isotopes – including 111In, 64Cu, 89Zr, and 68Ga – have been used for dosimetry imaging scans prior 90Y-based radiotherapy. However, these approaches leave much to be desired, as labeling a targeting vector (e.g., DOTATOC) with different radiometals can drastically change its in vivo properties [6–8]. Indeed, changes in the coordination environment, charge, or stability a radiometal–chelator complex [e.g., 90Y(DOTA)1− vs. 64Cu(DOTA)2−] can cause significant discrepancies in tumor uptake and organ distribution [9].
For this reason, the use of the positron-emitting radiometal 86Y (86Y, t1/2 = 14.7 h, β+ = 33%, keV) for PET imaging prior to 90Y-based therapy would appear to be ideal, as the 86Y- and 90Y-labeled radiopharmaceuticals would be chemically identical isotopologues [10]. Imaging with 86Y is a more attractive option than imaging with the commonly-used surrogate isotope 111In due to the former's suitability for PET (rather than SPECT) and its identical aqueous coordination chemistry to 90Y3+. However, the use of 86Y3+ nonetheless has its fair share of drawbacks. First, when compared to 90Y (t1/2 ~ 64.1 h), 86Y possesses a shorter half-life (14.7 h), which can preclude imaging studies at later time points. In addition, 86Y emits a relatively high-energy β+ (Emean = 640 keV), meaning that the positrons have a long mean-free path length (Rmean = 2.5 mm) compared to other popular positron-emitting radiometals such as 64Cu (Emean = 278.2 keV, Rmean = 0.56 mm) and 89Zr (Emean=402.7 keV, Rmean=1.27 mm) [11]. As a result of this higher β+ energy and an abundance of gamma emissions (1077, 1153, 1854, 1921 keV), the image quality of 86Y-PET scans is reported as being sub-optimal. However, the subtraction of some of these prompt gamma coincidences can improve image quality and useful images can be obtained from 86Y, despite the shortcomings described above [12]. Another concern that arises from this abundance of high-energy gamma emissions from 86Y is the radiation dose to anyone handling it. As a result, substantial radioactive shielding is required during radiolabeling, and the quantity of 86Y that can be transported in one container is limited [5]. For these reasons – as well as its limited availability in North America and the low purity of the available 86Y – experiments with 86Y were not included in this study. Although 86Y has previously been used successfully, the issues encountered with the available 86Y during this study prevented its inclusion [6,10,13–22].
Regardless of the identity of the metal, any radiopharmaceutical labeled with a metallic isotope must have a suitable chelator attached to the targeting vector (e.g., peptide, antibody) to facilitate the stable coordination of the radiometal ion [9,23–25]. The acyclic chelating ligand H4octapa (Fig. 1) has been previously synthesized and studied with the radiometals 111In (t1/2 ~ 2.8 days) and 177Lu (t1/2 ~ 6.6 days) and was observed to possess rapid radiolabeling kinetics and excellent in vitro and in vivo stability with both radiometals [26–29]. A popular class of targeting vectors to use with longer half-life radiometals such as 111In, 177Lu, and 90Y are antibodies, proteins that possess exquisite selectivity and binding affinity to their target antigens but are also sensitive to elevated temperatures (>40 °C). As a result, a chelator must be used that can coordinate radiometals at relatively low temperatures (e.g., 25–40 °C) so that the structural integrity of the antibody is not compromised. The acyclic chelator H4octapa was demonstrated to radiolabel with 111In and 177Lu in less than 15 min at room temperature. This suggests that radiolabeling performance with the chemically similar tricationic radiometals 86Y3+ and 90Y3+ should also be favorable.
Fig. 1.
The bifunctional chelators discussed in this work: p-SCN-Bn-H4octapa, p-SCN-Bn-CHX-A″-DTPA, and p-SCN-Bn-DOTA (C-DOTA).
One of the more common chelators used for 86Y/90Y is the macrocycle 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA; Fig. 1), which typically requires elevated temperatures of 60–90 °C over a period of 30–60 min for radiometalation [26–29]. The high temperatures required for radiolabeling make DOTA a very poor choice for use with antibodies. A newer and more suitable chelator for this radiometal pair is CHX-A″-DTPA, which – in contrast to DOTA – can be effectively radiolabeled at ambient temperatures within 15–30 min [26–31]. Although DOTA and CHX-A″-DTPA are the most established Y3+ chelators, a new NOTA derivative 3p-C-NETA has also been used with 177Lu, 86Y, and 90Y and gives high radiolabeling yields in only 5 min at room temperature [32–34].
H4octapa was previously studied with non-radioactive yttrium, with which it formed a complex with thermodynamic stability and solution chemistry comparable to the H4octapa complexes with Lu3+ and In3+[26,28,29]. These results with non-radioactive yttrium suggested that testing H4octapa with 90Y should yield fast radiolabeling kinetics and robust stability, as was found with 111In and 177Lu. To determine the suitability of H4octapa for use with isotopes of Y3+, the bifunctional chelators p-SCN-Bn-H4octapa and p-SCN-Bn-CHX-A″-DTPA were conjugated to the antibody trastuzumab and radiolabeled with 90Y, and the resulting radioimmunoconjugates were evaluated in vitro and in vivo. The stability of the 90Y chelate complexes were evaluated in vitro by serum stability and in vivo by measuring the uptake of 90Y in organs in which “free” 90Y3+ is known to accumulate (e.g., bone, liver, kidneys).
2. Materials and methods
2.1. General materials
All solvents and reagents were purchased from commercial suppliers (Sigma Aldrich, St. Louis, MO; TCI America, Portland, OR; Fisher Scientific, Waltham, MA) and were used as received unless otherwise indicated. DMSO used for chelator stock solutions was of molecular biology grade (>99.9%: Sigma, D8418). The bifunctional chelator p-SCN-Bn-H4octapa was synthesized as previously reported [28], and [(R)-2-amino-3-(4-isothiocyanatophenyl)propyl]-trans-(S,S)-cyclohexane-1,2-diamine-pentaacetic acid (p-SCN-Bn-CHX-A″-DTPA) and 1-(4-isothiocyanatophenyl)-3-[6,17-dihydroxy-7,10,18,21-tetraoxo-27-(N-acetylhydroxylamino)-6,11,17,22-tetraazaheptaeicosine] thiourea (p-SCN-Bn-DFO) were purchased from Macrocyclics, Inc. (Dallas, TX). UV–Vis measurements for determining antibody stock solution concentrations were taken on a Thermo Scientific Nanodrop 2000 spectrophotometer (Wilmington, DE), using the full-sized antibody (150 kDa) molar extinction coefficient of 210,000 L mol−1. 90YCl3 (0.5 M HCl) was purchased from PerkinElmer. 89Zr was produced at Memorial Sloan Kettering Cancer Center using an EBCO TR19/9 variable-beam energy cyclotron (Ebco Industries Inc., British Columbia, Canada) via the 89Y(p,n)89Zr reaction. 89Zr was purified in accordance with previously reported methods to create 89Zr with a specific activity of 5.3–13.4 mCi/μg (195–497 MBq/μg) [35]. Labeling reactions were monitored using silica–gel impregnated glass-microfiber instant thin layer chromatography paper (iTLC-SG, iTLC-SA, Varian, Lake Forest, CA) and analyzed on a Bioscan AR-2000 radio-TLC plate reader using Winscan Radio-TLC software (Bioscan Inc., Washington, DC). All radiolabeling chemistries were performed with ultra-pure water (>18.2 MΩ cm−1 at 25 °C, Milli-Q, Millipore, Billerica, MA) that had been stirred with Chelex-100 resin (1.2 g/L, 24 h) (BioRad Laboratories, Hercules, CA). Radioactivity in samples was measured using a Capintec CRC-15R dose calibrator (Capintec, Ramsey, NJ). For 89Zr biodistribution studies, a PerkinElmer (Waltham, MA) Automated Wizard Gamma Counter was used for counting radioactivity in organ samples and creating calibration curves for 89Zr. For 90Y biodistribution studies, a PerkinElmer (Waltham, MA) Automated Tri-Carb® 2910 TR liquid scintillation counter was used with samples that had been treated with Solvable™ (PerkinElmer) and Ultima Gold™ high flash-point universal liquid scintillation-cocktail (PerkinElmer).
2.2. Antibody modification
Trastuzumab (purchased commercially as Herceptin; Genentech, San Francisco, CA) was purified using PD10 size-exclusion columns [with phosphate buffered saline (PBS, pH 7.4), 3 times], followed by centrifugal filter units with a 50,000 molecular weight cut off (Amicon® ultra centrifuge filters, Ultracel®-50: regenerated cellulose, Millipore Corp., Billerica, MA) (PBS, pH 7.4) to remove α-α-trehalose dihydrate, L-histidine, and polysorbate 20 additives. After purification, the antibody was taken up in PBS pH 7.4 and kept in the fridge at 4 °C as a stock solution (12.8 mg/mL). Subsequently, 150 μL of antibody solution (~1.92 mg antibody) was combined with 850 μL PBS (pH 7.4), the pH of the resulting solution was adjusted to 8.8–9.0 with 0.1 M Na2CO3 (~30 μL), and 6 equivalents of p-SCN-Bn-H4octapa or p-SCN-Bn-CHX-A″-DTPA were added in 10–15 μL DMSO. The reactions were incubated at 37 °C for 1 h, followed by PD10 purification (PD10, Sephadex G-25 M, PD10 column, GE Healthcare; dead volume = 2.5 mL, product eluted with 2.0 mL fractions of sterile saline), and centrifugal filtration (Amicon 50 kDa) to purify the resultant antibody conjugate. The final immunoconjugate stock solutions were stored in PBS (pH 7.4) at 4 °C.
2.3. 90Y-radiolabeling of H4octapa- and CHX-A″-DTPA-trastuzumab
Aliquots of chelate-modified antibody (200 μg for 90Y) bearing either H4octapa or CHX-A″-DTPA were transferred to 2 mL microcentrifuge tubes and made up to 0.5 mL with ammonium acetate buffer (pH 5.5, 200 mM, treated with Chelex-100 resin at 1.2 g/L overnight before use). Aliquots of 90Y (~0.35 mCi, ~12.9 MBq) were added. The radiolabeling mixtures were allowed to react at 37 °C (500 rpm, thermomixer) for 60 min and then analyzed via iTLC-SA (acidic) with an eluent of 50 mM DTPA (pH 2.5). Crude radiochemical yields for 90Y-CHX-A″-DTPA-trastuzumab were ~96% and for 90Y-octapa-trastuzumab were ~99%. After 60 min, EDTA solution was added to quench the reactions (20 μL EDTA, 50 mM, pH 5.5), and were then purified using size-exclusion chromatography (PD10), followed by centrifugal filtration (Amicon® ultra 50 kDa, rinsed 3 times with saline). The radiochemical purity of the final purified radiolabeled bioconjugate was assayed by radio-iTLC-SA (DTPA mobile phase, 50 mM, pH 2.5). For 90Y reactions, crude radiochemical yields of both compounds were >95%, and purified yields were all >99%. In the iTLC experiments, 90Y-octapa-trastuzumab and 90Y-CHX-A″-DTPA-trastuzumab remained at the baseline, while 90Y3+ ions eluted with or near the solvent front (~100–150 mm) with the DTPA mobile phase (50 mM, pH 2.5).
2.4. 89Zr-DFO-trastuzumab radiolabeling
89Zr-DFO-trastuzumab was prepared as previously published, and p-SCN-Bn-DFO was conjugated to trastuzumab using the same method as described above for p-SCN-Bn-H4octapa and p-SCN-Bn-CHX-A″-DTPA.[36,37] Briefly, aliquots of 89Zr oxalate were neutralized to ~pH 7.0–7.4 using sodium carbonate (1 M), and subsequently mixed with DFO-trastuzumab (~500 μg, 0.5 mL PBS, pH 7.4) and reacted for 60 min at 37 °C. Radiochemical yields obtained were >99% after 1 h, and 89Zr-DFO-trastuzumab was purified as described above for the 90Y-labeled radioimmunoconjugates.
2.5. MALDI-TOF MS/MS analysis to determine the number of chelates per antibody
The number of benzylthiourea-linked H4octapa or CHX-A″-DTPA chelates conjugated to trastuzumab was determined using MALDI-TOF MS/MS (Alberta Proteomics and Mass Spectrometry Facility, University of Alberta, Canada). All experiments were performed in triplicate, and all samples were run along with standard samples of unmodified trastuzumab (run on the same day). 1 μL of each sample (1 mg/mL) was mixed with 1 μL of sinapic acid (10 mg/mL in 50% acetonitrile:water and 0.1% trifluoroacetic acid). 1 μL of the sample/matrix solution was then spotted onto a stainless steel target plate and allowed to air dry. All mass spectra were obtained using a Bruker Ultraflex MALDI-TOF/TOF (Bruker Daltonic GmbH). Ions were analyzed in positive mode, and external calibration was performed using of a standard protein mixture (bovine serum albumin). The mass signals (M+2/2) at half the molecular weight of the antibody were taken from each chromatogram and averaged (n = 3), and the molecular weight of the unmodified trastuzumab was subtracted from the molecular weight of the modified antibody to determine the mass contributed by the conjugated chelators. The mass difference was divided by the molecular weight of the attached bifunctional chelator (p-SCN-Bn-H4octapa, p-SCN-Bn-CHX-A″-DTPA), and the error in each triplicate set of measurements (unmodified trastuzumab vs. modified trastuzumab) was propagated to the final value of the number of chelates per antibody. The MALDI-TOF spectra are available in the supporting information Figs. S1–S9.
2.6. Determination of immunoreactivity
The immunoreactivity of 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab was determined using specific radioactive cellular-binding assays in which an excess of cells was used (~10 million per microcentrifuge tube). This quantity of cells was assumed to contain effectively “infinite antigen” relative to the quantity of antibody added to each (~ 6 ng of radiolabeled antibody). To this end, 10 million SKOV3 cells were suspended in microcentrifuge tubes in ~0.2 mL of media in microcentrifuge tubes. Aliquots of the bioconjugates – 90Y-octapa-trastuzumab and 90Y-CHX-A″-DTPA-trastuzumab (20 μL of a stock solution containing ~10–20 μCi, ~0.37–0.73 MBq, ~3 μg of radioimmunoconjugate, in 10 mL of 1% bovine serum albumin in PBS pH 7.4) – were added to each tube and mixed by pipette (n = 4 each immunoconjugate; final volume: ~220 μL) The samples were then left to sit on ice for 60 min. Subsequently, the treated cells were pelleted via centrifugation (600 g, 4 °C, for 2 min), and the media was transferred by pipette to clean microcentrifuge tubes. The cell pellets were not re-suspended and were washed three times with ice-cold PBS (1 mL) and centrifuged (600g, 4 °C, 2 min) between each wash. Each wash was collected into an empty microcentrifuge tube (1 mL). Each tube (cell pellet, media, and 3 PBS washes) was measured in a scintillation counter (other isotopes can be counted using an automated gamma counter). Each tube was rinsed with 0.5 mL of PBS and transferred to scintillation vials (5 mL), 4.5 mL of Ultima gold scintillation fluid was added, samples were vortex mixed for 30 s, and then measured on an automated scintillation counter (PerkinElmer). The activity data were background-corrected, and the amount of activity (antibody) bound to the cells was compared to the total amount of activity present in the media and washes. The immunoreactivity was determined by dividing the amount of radioactivity bound to the cell pellet by the total amount of radioactivity present in the cell pellet, the media, and the washes.
2.7. 90Y-octapa-trastuzumab and 90Y-CHX-A″-DTPA-trastuzumab blood serum competition experiments
Frozen human blood serum was thawed for 30 min, and 500 μL aliquots were transferred to 2.0 mL Corning microcentrifuge vials. A portion of radiolabeled immunoconjugate (~150 μCi, ~50–75 μg, 500 μL, purified) was transferred to the blood serum (n=3 for each chelator). Serum competition samples were then incubated at 37 °C with gentle agitation (500 rpm, Eppendorf thermomixer) and analyzed via iTLC-SA elution with a DTPA eluent (50 mM, pH 2.5) followed by scanning (Bioscan AR-2000) at time points of 0, 1, 24, 48, 72, and 96 h.
2.8. Cell culture
Human ovarian cancer cell line SKOV3 was obtained from the American Tissue Culture Collection (ATCC, Bethesda, MD) and maintained in a 1:1 mixture of Dulbecco's modified Eagle medium: F-12 medium, supplemented with 10% heat-inactivated fetal calf serum (Omega Scientific, Tarzana, CA), 2.0 mM glutamine, nonessential amino acids, 100 units/mL penicillin, and 100 units/mL streptomycin in a 37 °C environment containing 5% CO2. Cell lines were harvested and passaged weekly using a formulation of 0.25% trypsin/0.53 mM EDTA in Hank's Buffered Salt Solution without calcium and magnesium.
2.9. Subcutaneous SKOV3 xenografts
All experiments were performed under an Institutional Animal Care and Use Committee-approved (IACUC) protocol, and the experiments followed institutional guidelines for the proper and humane use of animals in research. Six- to eight-week-old athymic nu/nu female mice (NCRNUM) were obtained from Charles River Laboratories (Kingston, NY). Animals were housed in ventilated cages, were given food and water ad libitum, and were allowed to acclimatize for approximately 1 week prior to treatment. SKOV3 tumors were induced on the left shoulder by a subcutaneous injection of 5.0 × 106 cells in a 100 μL cell suspension of a 1:1 mixture of fresh media/BD Matrigel (BD Biosciences, Bedford, Ma). Experiments were performed 3–4 weeks following the injection of the cancer cells.
2.10. 90Y-octapa-trastuzumab and 90Y-CHX-A″-DTPA-trastuzumab biodistribution studies
The purified 90Y immunoconjugates (radiolabeled and purified as described above) were suspended in sterile saline (0.9% NaCl) to a concentration of ~20–25 μCi (1.1 MBq, ~ 15–20 μg) in 200 μL. The specific activity of both 90Y-octapa-trastuzumab and 90Y-CHX-A″-DTPA-trastuzumab was ~1.8 mCi/mg. Biodistribution experiments could not be performed using a gamma counter for 90Y, as the strictly β−-emitting isotope requires organ digestion, bleaching, and scintillation counting. After doses were drawn in syringes, they were weighed before and after injection to obtain the total mass of injected antibody solution. This weight was then used with a set of standards to determine the total injected dose in each mouse in scintillation counts (in counts per mg of injected dose; see below).
Each mouse was injected with 90Y-trastuzumab via the tail vein and euthanized by CO2 (g) asphyxiation at time points of 24 and 72 h after the administration of the radioimmunoconjugate (n = 4 per time point). After sacrifice, the blood, tumor, heart, lungs, liver, spleen, pancreas, kidneys, large intestine, small intestine, muscle, bone (femur), and skin (ear) were collected. All organs were rinsed in water after removal and air-dried for 5 min. The organs were collected into pre-weighed glass scintillation vials (20 mL), and samples of well-vascularized tissues with higher blood content – including the blood, spleen, and liver – were obtained in small quantities (50–100 mg) to minimize color quenching in the final digested organ solutions. Subsequently, tissues were weighed and then digested for scintillation counting. Full tables of biodistribution data for 90Y (%ID/g values), and experimental details for organ digestion are available in the supporting information Tables S1–S4.
A control biodistribution experiment using unbound 90YCl3 was performed as well. In this experiment, an aliquot of pure 90YCl3 (~100 μCi) was transferred to sterile saline (1.0 mL), and doses were drawn (~20 μCi in 200 μL) for injection (n = 3). In this case, the biodistribution experiment was performed only a 72 h post-injection. The organ samples were weighed and measured by scintillation counting as described above.
Standards for the 90Y biodistribution experiments were prepared by transferring a small volume of the same injected 90Y-antibody or 90YCl3 doses in saline (in the same formulations as were injected into mice, ~20 μL, ~2 μCi) into pre-weighed glass scintillation vials (20 mL vials, same as used for biodistributions, n = 5) and then weighing the mass of the 20 μL transferred mixture. These standards were then processed in the same manner as described above for organ samples and counted on the scintillation counter at the same time as the organ samples. The average number of scintillation counts per milligram of standard injected dose was calculated from these standards (counts per milligram), and the mass of injected dose for each biodistribution (obtained by subtracting weight of empty syringes from full syringes after injection) was then multiplied by this standard factor to obtain a value for the total injected dose for each mouse in scintillation counts.
2.11. 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab preliminary radioimmunotherapy pilot study
Female nude mice (n = 24) were xenografted with SKOV3 cells (subcutaneous, left shoulder) as described above. Tumor growth and volume were monitored by caliper measurements, in which the length (L) was taken as the longest dimension, and the width (W) as the shortest. Tumor volume (V) was calculated using the formula [V = L × (W/2)2]. Once the tumors grew to 50–100 mm3, the mice with the most consistent tumors were arranged in cages of 4 with approximately equal average tumor volumes amongst all mice. Mice were arranged into 3 groups (n = 4 each) using ear tags and ear punching as methods of sorting and identification. Tumors in several mice did not take, and some tumor sizes were very inconsistent, so smaller groups of n = 4 were used for this preliminary radioimmunotherapy pilot study (n = 5 for controls). Groups were injected with 90Y-octapa-trastuzumab (n = 4, ~100 μCi, 200 μL saline, ~30 μg antibody, ~3.3 mCi/mg SA), 90Y-CHX-A″-DTPA-trastuzumab (n = 4, ~100 μCi, 200 μL saline, ~30 μg antibody, ~3.3 mCi/mg), and controls (n = 5, 200 μL saline, ~30 μg trastuzumab antibody). New batches of CHX-A″-DTPA-trastuzumab and octapa-trastuzumab were synthesized to preclude aggregation and abnormal spleen uptake similar to that observed in the biodistribution of 90Y-octapa-trastuzumab. Mice were assessed via tumor caliper measurements and bodyweight twice per week for the duration of the experiment, which was terminated when tumors approached the size limit of 1000 mm3. At the termination of the 90Y therapy study (day 36), mice were injected with 89Zr-DFO-trastuzumab (~130 μCi, ~30 μg) to allow for PET imaging, biodistribution, and autoradiography/histology experiments. These experiments could not be performed with the strictly β−-emitting isotope 90Y (due both to the 36 day therapy study and 90Y's lack of analytically useful gamma or positron emissions).
2.12. Cerenkov luminescence imaging (CLI)
Optical images of Cerenkov radiation (CR) were obtained using an IVIS 200 (Caliper Life Sciences) optical imaging machine. This system uses a cryo-cooled charge-coupled device for high-sensitivity detection of low-intensity sources. The mice that had been injected with 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab for the radioimmunotherapy experiments (n = 4 per group; ~100 μCi/mouse) were imaged using two-minute exposures 72 h after the administration of the therapeutic radioimmunoconjugates.
2.13. 89Zr-DFO-trastuzumab PET imaging and biodistribution studies
Mice from the 90Y therapy study (36 days post-injection of 90Y or control) were imaged via PET with 89Zr-DFO-trastuzumab (n = 4 from each therapy group). To this end, 89Zr-DFO-trastuzumab was radiolabeled, purified, and prepared as described above. PET imaging was performed using a micro-PET R4 rodent scanner (Focus 120, Concord Microsystems). Mice were administered radiolabeled antibody (~30 μg, ~130 μCi, ~4.8 MBq) in 200 μL sterile saline (0.9% NaCl) via intravenous tail vein injection. Approximately 5 min prior to PET image acquisition, mice were anesthetized via inhalation of 2% isoflurane/oxygen gas mixture (Baxter Healthcare, Deerfield, IL) and placed on the scanner bed where anesthesia was maintained. PET images were acquired at 72 h, with PET data being recorded via static scans with a minimum of 15 million coincident events (~30 min). An energy window of 350–700 keV and a coincidence timing window of 6 ns was used. Data were sorted into 2D histograms by Fourier rebinning, and transverse images were reconstructed by filtered back-projection (FBP) into a 128 × 128 × 63 (0.72 × 0.72 × 1.3 mm3) matrix. The image data were normalized to correct for non-uniformity of response of the PET, dead-time count losses, positron branching ratio, and physical decay to the time of injection, but no attenuation, scatter, or partial-volume averaging correction was applied. The counting rates in the reconstructed images were converted to activity concentrations (percentage injected dose [%ID] per gram of tissue) by use of a system calibration factor derived from the imaging of a mouse-sized water-equivalent phantom containing 89Zr. Images were analyzed using ASIPro VM software (Concorde Microsystems).
These 89Zr-DFO-trastuzumab-treated mice were euthanized for the biodistribution experiments immediately following the single 72 h PET imaging time point. Biodistribution experiments with 89Zr-DFO-trastuzumab did not require organ digestion or scintillation counting, as the gamma emissions from 89Zr could be quantified using an automated gamma counter. The organs from 89Zr biodistribution experiments were placed in pre-weighed test tubes, weighed, and then the amount of radioactivity present was determined using automated gamma counting (PerkinElmer). The counts were background- and decay-corrected from the time of injection and then converted to the percentage of injected dose (%ID) per gram of organ tissue (%ID/g). The radioactivity counts measured in each organ were converted to activity (μCi) using a calibration curve created from known standards of 89Zr (serial dilution from ~3 μCi 89Zr). Biodistribution data from mice injected with 89Zr-DFO-trastuzumab can be found in the supporting information (Tables S5–S7).
2.14. Autoradiography and histology of 89Zr-DFO-trastuzumab in 90Y treated tumors
Following PET imaging, tumors were excised and embedded in optimal-cutting-temperature mounting medium (OCT, Sakura Finetek) and frozen on dry ice. Series of 10 μm frozen sections were then cut. To determine radiotracer distribution, digital autoradiography was performed by placing tissue sections in a film cassette against a phosphor imaging plate (Fujifilm BAS-MS2325; Fuji Photo Film) for an appropriate exposure period at −20 °C. Phosphor imaging plates were read at a pixel resolution of 25 μm with a Typhoon 7000 IP plate reader (GE Healthcare). After autoradiographic exposure, the same frozen sections were then used for immunohistochemical (IHC) staining and microscopy. The immunohistochemistry detection of Androgen Receptor was performed using a Discovery XT processor (Ventana Medical Systems). A rabbit polyclonal C-erbB2 antibody (Enzo, cat.# ALX-810-227) was used at 10 μg/mL concentration. The tissue sections were fixed in 4% paraformaldehyde and blocked for 30 min in 10% normal goat serum, 2% BSA in PBS. The incubation with the primary antibody was done for 5 h, followed by 16 min incubation with biotinylated goat anti-rabbit IgG (Vector labs, cat#:PK6101) in 1:200 dilution. Blocker D, streptavidin–HRP and DAB detection kit (Ventana Medical Systems) were used according to the manufacturer instructions. Sequential sections were stained with H&E. Whole mount images were acquired at ×100 magnification using a BX60 microscope (Olympus America, Inc.) equipped with a motorized stage (Prior Scientific Instruments Ltd.) and DP80 camera (Olympus). Whole-tumor montage images were obtained by acquiring multiple fields at ×40 magnification, followed by alignment using MicroSuite Biologic Suite (version 2.7; Olympus). IHC and autoradiographic images were registered using Adobe Photoshop (CS6) as previously described [38].
3. Results and discussion
3.1. H4octapa coordination chemistry
Solution nuclear magnetic resonance (NMR) data and density functional theory (DFT) calculations from a previous study exploring the solution chemistry of H4octapa with Y3+ suggested that this metal–ligand pair forms complexes similar to those of H4octapa with In3+ and Lu3+[26]. More specifically, the structures of [Y(octapa)]− and [Lu(octapa)]− were predicted for both 8-coordinate (ligand only) and 9-coordinate (ligand and one H2O) architectures and showed striking similarities. Furthermore, the molecular electrostatic potentials (MEP) of the [Y(octapa)]− and [Lu(octapa)]− complexes were mapped onto the surface of the DFT-calculated structures, suggesting very similar charge distributions between the two complexes [26,28]. Potentiometric titrations determined H4octapa to have a formation constant (log KML) of 18.3 ± 0.1 with Y3+ and 20.08 ± 0.9 with Lu3+, revealing high thermodynamic stability [26,28]. Another previous study compared the in vivo performance of H4octapa-trastuzumab and DOTA-trastuzumab with the radiometals 111In and 177Lu, highlighting the improved radiolabeling properties of H4octapa compared to DOTA [28]. This preliminary in vivo work demonstrated that H4octapa is a strong chelator for the radiometals 111In and 177Lu, and the similarities in non-radioactive characterization between the [Y(octapa)]− and [Lu(octapa)]− complexes suggest that H4octapa may be an excellent ligand for radiopharmaceutical applications with 86Y and 90Y. The chelator H4octapa presents an alternative to the “gold standard” chelator CHX-A″-DTPA for radiometals such as 111In, 177Lu, and 86Y/90Y.
3.2. Bioconjugation, radiolabeling, and in vitro characterization
The purpose of this study was to evaluate the in vitro and in vivo performance of the chelator H4octapa with the radiometal 90Y and compare it with the existing `gold standard' yttrium chelator CHX-A″-DTPA. Although DOTA forms superbly stable complexes with radiometals such as 111In, 177Lu, and 86Y/90Y, its radiolabeling kinetics are very slow, and it the high temperature conditions that it requires for radiolabeling are not compatible with antibodies. To begin, trastuzumab (HER2-targeting antibody, ~150 kDa) was incubated under slightly basic conditions (pH 8.5–9.0) with 6 equivalents of p-SCN-Bn-H4octapa or p-SCN-Bn-CHX-A″-DTPA and purified via size exclusion chromatography (2 × PD10) and spin filtration (2 × Amicon Ultra 50 kDa). After purification, aliquots of trastuzumab immunoconjugates (10 μL, 1 μg/μL, n = 3) were frozen and sent for MALDI-TOF analysis. Mass spectrometry results indicated that these modifications yielded 1.5 ± 0.1 chelates per antibody in the case of p-SCN-Bn-H4octapa and 1.0 ± 0.6 chelates per antibody in the case of p-SCN-Bn-CHX-A″-DTPA.
H4octapa-trastuzumab and CHX-A″-DTPA-trastuzumab were radiolabeled with 90Y3+ in NH4OAc buffer (pH 5.5, 200 mM, treated with Chelex-100 resin) for 60 min at 37 °C. Radiolabeling yields were evaluated using iTLC-SA (acidic) silica-embedded paper strips, and a mobile phase of DTPA (pH 2.5, 50 mM) was used to resolve “free” 90Y3+ from antibody-bound radiometal (Fig. 2). High radiochemical yields were easily achieved with 90Y3+ (>95% radiochemical yield crude, >99% purity after purification). Both H4octapa-trastuzumab and CHX-A″-DTPA-trastuzumab (50 μg) were also radiolabeled with 90Y at room temperature to compare their kinetics. In these experiments, the radiochemical yields were evaluated after 15 min, with both immunoconjugates obtaining crude radiochemical yields of >95% (by iTLC), revealing equally facile radiolabeling kinetics with 90Y.
Fig. 2.
Radiolabeling yields with 90Y as determined by iTLC (iTLC-SA acidic strips, mobile phase 50 mM DTPA, pH 2.5). Un-coordinated 90Y3+ elutes with the solvent front of the iTLC-SA strip (~100–125 mm), while the 90Y-labeled radioimmunoconjugate remains at the baseline (~25 mm).
The radiolabeled immunoconjugates were evaluated by a cell-binding assay to determine their immunoreactivity (Table 1, % of native binding affinity). This in vitro assay used the same SKOV3 cancer cells used for in vivo experiments. Briefly, the antigen over-expressing cells (10 million SKOV3 per microcentrifuge tube [39]) were mixed with a small quantity of radioimmunoconjugate (~6 ng), and the percent radioimmunoconjugate bound to cells after washing was determined as the % immunoreactivity. The immunoreactivity values for 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab were 93.9 ± 0.9% and 91.2 ± 0.8%, respectively. These values indicate that chelator conjugation and radiolabeling yielded the final radioimmunoconjugates with minimal antibody degradation or radiolysis, and did not disrupt their ability to bind their target antigen. In order to test the stability of the radioimmunoconjugates under biologically relevant conditions, both constructs were incubated in human serum for 3 days at 37 °C. At the end of the experiment (96 h), the stability of the 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab conjugates were found to be excellent: 87.1 ± 0.9% and 94.8 ± 0.6%, respectively (Table 1, Fig. 3). In summary, the radiolabeling yields, radiochemical purity, immunoreactivity, and serum stability results were excellent for both 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab. Despite the problems encountered with 86Y that precluded its inclusion in this study, 86Y remains an attractive and potentially useful isotope for PET imaging applications.
Table 1.
Chemical and in vitro biological characterization data for 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab radioimmunoconjugates.
Chelator | Isotope | Radiolabeling yields (37 °C, 60 min) | Chelates/mAba | Specific activity (mCi/mg) | Immunoreactive fraction (%)b | Serum stability 96 h (%)c |
---|---|---|---|---|---|---|
H4octapa | 90Y | Crude = 99%, Pure = 99% | 1.5 ± 0.1 | 1.8 | 91.2 ± 0.8 | 94.8 ± 0.6 |
CHX-A″-DTPA | 90Y | Crude = 96%, Pure = 99% | 1.0 ± 0.6 | 1.8 | 93.9 ± 0.9 | 87.1 ± 0.9 |
MALDI-TOF MS/MS mass spectrometry (n = 3) spectra in Figs. S1–S9.
Determined by saturation binding cell assay (10 × 106 SKOV3 cells per vial, n = 4).
Determined by incubation in human serum (37 °C for 96 h; n = 3).
Fig. 3.
Stability comparison of 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab in human blood serum from 0 to 96 h (37 °C, 500 rpm mixing), showing excellent stability for both radioimmunoconjugates (87.1 ± 0.9% and 94.6 ± 0.3% at 96 h, respectively).
3.3. Biodistribution studies of 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab
To expand further our comparison of the new chelator H4octapa to the incumbent CHX-A″-DTPA, the in vivo performance of the 90Y-labeled immunoconjugates was assessed via biodistribution experiments in a model system using HER2-expressing SKOV3 ovarian cancer cells. Each of the radiolabeled antibodies – 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab – was injected via tail vein into female nude athymic mice bearing subcutaneous SKOV3 ovarian cancer xenografts on the left shoulder (~20–30 μCi, 0.74–1.1 MBq, ~15–20 μg, in 200–250 μL of sterile saline; tumor volume ~ 100–150 mm3). After 24 and 72 h (n = 4 per time point), the mice were euthanized via CO2 (g) asphyxiation, and 14 organs including the SKOV3 tumors were removed, weighed, and assayed for radioactivity content on a scintillation counter (digested and bleached organ samples). A control biodistribution experiment was also performed with 90YCl3, which illustrated that “free” 90Y3+ accumulates primarily in the lungs, liver, spleen, kidneys, and bones.
The biodistribution profiles of both 90Y-labeled immunoconjugates were nearly identical with the exception of higher uptake of 90Y-octapa-trastuzumab in the spleen and slightly higher tumor uptake with 90Y-CHX-A″-DTPA-trastuzumab. Higher spleen uptake is typically a result of antibody aggregation, which can occur during the storage, radiolabeling, purification, or formulation of radiolabeled antibodies. The slightly lower tumor uptake observed for 90Y-octapa-trastuzumab could be because part of the injected dose was trapped in the spleen (possibly due to aggregation) instead of being free to accumulate in the tumor. The uptake of both radioimmunoconjugates was identical (within error) in organs that are known to accumulate free 90Y3+ (e.g., kidneys, liver, bone), which suggests that the higher spleen uptake observed for 90Y-octapa-trastuzumab was not a result of chelate instability. The control group of 90YCl3 clearly demonstrates which organs “free” 90Y accumulates in, suggesting that both chelators offer equivalent and impressive in vivo stability (Fig. 4). Importantly, subsequent radioimmunotherapy experiments performed with 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab (vide infra) showed no signs of abnormal spleen uptake for either tracer. Antibody aggregation sometimes occurs during radiolabeling, purification, and handling, and it appears that it did not occur for this 90Y therapy study. This comparison suggests that both H4octapa and CHX-A″-DTPA are highly stable and appropriate choices for 90Y chelation for in vivo use.
Fig. 4.
Biodistribution data showing a comparison of the stability and performance of 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab (S.I.=small intestine, L.I.=large intestine), including a control group inject with 90YCl3 used to demonstrate the primary organs in which “free” 90Y3+ accumulates.
3.4. 90Y Radioimmunotherapy and Cerenkov luminescence imaging using 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab in mice bearing subcutaneous SKOV3 xenografts
After the successful radiolabeling and biodistribution results with 90Y described above, it was deemed prudent to test these two radioimmunoconjugates for their practical ability to deliver therapeutic doses of 90Y to target tissue. To this end, therapeutic doses (100 μCi, 3.7 MBq) of both 90Y-labeled radioimmunoconjugates were injected into cohorts of mice bearing subcutaneous SKOV3 xenografts. More specifically, three cohorts of mice were prepared (n = 4 each treatment, n = 5 control): 90Y-CHX-A″-DTPA-trastuzumab (30 μg, 100 μCi, 3.7 MBq), 90Y-octapa-trastuzumab (30 μg, 100 μCi, 3.7 MBq), and controls (30 μg non-radiolabeled trastuzumab). Cerenkov luminescence imaging (CLI) was performed 3 days after the injection of the radioimmunoconjugates, which provided visual confirmation of tumor uptake that could not be obtained by PET or SPECT imaging with 90Y (Fig. 5). To ensure that no abnormal uptake in the spleen was observed for either antibody (typically the result of antibody aggregation during radiolabeling and the formulation of doses), the mice were also imaged by CLI on the left side (the side opposite the subcutaneous SKOV3 xenografts). The spleen sits close to the skin on the left side of mice, and therefore high spleen uptake should be observable by CLI. No accumulation in the spleen could be observed by CLI in this experiment, suggesting no abnormal uptake in the spleen.
Fig. 5.
Cerenkov luminescence imaging of 90Y therapy mice 72 h after the administration of 90Y-CHX-A″-DTPA-trastuzumab or 90Y-octapa-trastuzumab (100 μCi per mouse, ~30 μg trastuzumab, 2-min imaging time), anesthetized with isoflurane, imaged for 2 min, showing substantial accumulation in the HER2-expressing SKOV3 tumors.
The tumor volumes of the 90Y-treated mice were monitored twice per week using caliper measurements, and the general health of the mice was observed by appearance and body weight. This preliminary radioimmunotherapy pilot study was started with larger cohorts of mice bearing SKOV3 xenografts (n=8 per group) to account for tumors that did not take and for tumors that were much larger or smaller than average. This approach yielded cohorts of n=4 with similarly sized tumors for each therapy group and n=5 for the control group. By day 36, the control tumors were approaching the 1000 mm3 limit of the study, so this time was chosen as the end point for the study. A plot of the normalized tumor growth vs. time is displayed in Fig. 6 and clearly shows nearly identical treatment efficacy for the 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab groups, with substantial reductions in tumor growth rate being compared to the control group. This demonstrates that the serum stability and biodistribution experiments – which suggested comparable stability and in vivo performance of 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab – did indeed translate to equivalent radioimmunotherapeutic efficacy as measured by tumor growth response. The slightly lower tumor uptake observed in the biodistribution study of 90Y-octapa-trastuzumab could have been a result of higher spleen uptake due to a minor amount of aggregation of the antibody during radiolabeling, which was not observed during the 90Y therapy study. These results confirm that the chelators H4octapa and CHX-A″-DTPA are effective choices for 90Y in terms of radiolabeling kinetics, stability, and in vivo performance.
Fig. 6.
Normalized tumor growth during the 36 day therapy experiment, showing substantial reductions in tumor growth in the cohorts treated with 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab compared to the control group that was treated with trastuzumab only.
3.5. Post-radioimmunotherapy 89Zr-DFO-trastuzumab PET imaging, biodistribution, and autoradiography in mice following radioimmunotherapy
PET scans are commonly used in the clinic to monitor the response of cancer patients to therapy. However, in the case of this radioimmunotherapy study, the therapeutic isotope (90Y) does not emit any particles suitable for imaging. Furthermore, 36 days after the administration of the 90Y-labeled radioimmunoconjugates in this study, too little radioactive 90Y was left to perform autoradiography experiments. At the conclusion of the therapy study, the treated mice were injected with 89Zr-DFO-trastuzumab to facilitate PET imaging and autoradiography of tumor sections. To this end, at the end of the 90Y radioimmunotherapy study (day 36), the mice from each group (n = 4) were injected with 89Zr-DFO-trastuzumab (~130 μCi, ~4.8 MBq, ~30–35 μg) and imaged after 72 h. The PET images in Fig. 7 illustrate substantial uptake of 89Zr-DFO-trastuzumab in the SKOV tumors of the mice from the 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab radioimmunotherapy therapy cohorts. Not surprisingly, these images provide a telling picture of the substantially larger tumors of the animals in the control group. Furthermore, lower activity concentrations of 89Zr-DFO-trastuzumab were observed in tumors of the control group, most likely due to the large size of the tumors (~1000 mm3) and the presence of poorly-perfused necrotic regions.
Fig. 7.
89Zr-DFO-trastuzumab PET imaging of representative mice from the 90Y radioimmunotherapy experiment. The mice were injected with 89Zr-DFO-trastuzumab (~30 μg, ~130 μCi, ~4.8 MBq) on day 36 of the radioimmunotherapy study and imaged 72 h later. The images clearly show higher uptake of 89Zr-DFO-trastuzumab in the tumors of the mice that had been treated with 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab and lower uptake in the much larger tumors of the mice from the control group.
The localization of 89Zr-DFO-trastuzumab in the mice from the therapy study was evaluated via biodistribution, which showed similar tumor uptake and tissue distribution in both therapy groups. Tumor uptake in control mice was lower than therapy groups as a result of the substantially larger tumors (expressed as %ID/g). During the biodistribution necropsy (Fig. 8), pieces of SKOV3 tumor were taken from the 90Y treated and control mice that had been injected with 89Zr-DFO-trastuzumab. Autoradiography and histology (hematoxylin and eosin as well as HER2 staining) were performed on sections of tumors from each therapy group (Fig. 9). Results confirm high expression of HER2 in SKOV3 tumors, with the uptake of 89Zr-DFO-trastuzumab mimicking the distribution that would be expected from the 90Y-labeled radioimmunoconjugates. Interestingly, the distribution of 89Zr-DFO-trastuzumab in the tumors of the 90Y-octapa-trastuzumab therapy group (Fig. 9, row 2) shows high concentrations of 89Zr in a region with low HER2 expression. This is most likely a result of fluid pooling in a necrotic region of the tumor, which likely formed as a result of radioimmunotherapy. 89Zr-DFO-trastuzumab showed similar uptake in both 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab groups (Figs. 7 and 8).
Fig. 8.
Biodistribution of 89Zr-DFO-trastuzumab in the mice from the 90Y radioimmunotherapy experiment. The mice were injected with 89Zr-DFO-trastuzumab on day 36 of the radioimmunotherapy trial and euthanized for biodistribution 72 h after the administration of the imaging agent.
Fig. 9.
Sections of tumors from the 89Zr-trastuzumab PET/biodistribution obtained at the conclusion of the 90Y radioimmunotherapy trial, showing representative SKOV3 tumors from the (1) 90Y-CHX-A″-DTPA-trastuzumab group, (2) 90Y-octapa-trastuzumab group, and (3) control (trastuzumab only) group. Panels display (A) hematoxylin- and eosin (H&E)-stained tumor sections outlining the architecture of the tumor; (B) immunofluorescence staining of HER2 expression; (C) digital autoradiography demonstrating the distribution of 89Zr-DFO-trastuzumab within the tumors; and (D) overlay of the digital autoradiograph with the H&E and HER2 staining.
4. Conclusions
The principal benefit of acyclic chelators such as H4octapa and CHX-A″-DTPA over macrocyclic chelators such as DOTA lies in the ability of the former to be radiolabeled with rapid kinetics without significant heating. Radiolabeling experiments revealed that both CHX-A″-DTPA-trastuzumab and H4octapa-trastuzumab could be labeled with 90Y3+ in very high radiochemical purity after only 15 min at room temperature. Both 90Y-CHX-A″-DTPA-trastuzumab and 90Y-octapa-trastuzumab exhibited excellent in vitro and in vivo stability, high immunoreactivity, and comparable biodistribution profiles in mice bearing HER2-expressing SKOV3 ovarian cancer xenografts. Furthermore, both 90Y-labeled radioimmunoconjugates displayed identical therapeutic efficacy in radioimmunotherapy studies in mice bearing SKOV3 tumors. In these experiments, 36 days after the administration of the radioimmunoconjugates, the average tumor volume in both treatment groups was ~300 mm3, a remarkable shift compared to the average volume of ~1000 mm3 for the tumors of the control group. In the end, these data clearly suggest that H4octapa and CHX-A″-DTPA are both suitable chelators for use with 90Y.
Supplementary Material
Acknowledgments
Eric W. Price was supported by a Natural Sciences and Engineering Research Council (NSERC) postdoctoral fellowship (PDF, 2014–2016). The MSKCC Small-Animal Imaging Core Facility, supported in part by NIH Small-Animal Imaging Research Program (SAIRP) Grant No. R24 CA83084 and NIH Center Grant No. P30 CA08748, is gratefully acknowledged. The Radiochemistry and Molecular Imaging Probe core is also supported in part by NIH Center grant N0P30CA08748. The authors also thank the NIH (Award 4R00 CA178205-2, B.M.Z.).
Footnotes
These authors declare no competing financial interests.
Ethical approval All applicable institutional and national guidelines for the care and use of animals were followed.
Appendix A. Supplementary data Supplementary data to this article can be found online at http://dx.doi.org/10.1016/j.nucmedbio.2016.06.004.
References
- [1].Forrer F, Valkema R, Kwekkeboom DJ, de Jong M, Krenning EP. Peptide receptor radionuclide therapy. Best Pract Res Clin Endocrinol Metab. 2007;21:111–29. doi: 10.1016/j.beem.2007.01.007. [DOI] [PubMed] [Google Scholar]
- [2].Larson SM, Carrasquillo JA, Cheung N-KV, Press OW. Radioimmunotherapy of human tumours. Nat Rev Cancer. 2015;15:347–60. doi: 10.1038/nrc3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Wadas TJ, Wong EH, Weisman GR, Anderson CJ. Coordinating Radiometals of copper, gallium, indium, yttrium, and zirconium for PET and SPECT imaging of disease. Chem Rev. 2010;110:2858–902. doi: 10.1021/cr900325h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Forrer F, Waldherr C, Maecke HR, Mueller-Brand J. Targeted radionuclide therapy with 90Y-DOTATOC in patients with neuroendocrine tumors. Anticancer Res. 2006;26:703–7. [PubMed] [Google Scholar]
- [5].Holland JP, Williamson MJ, Lewis JS. Unconventional nuclides for radiopharmaceuticals. Mol Imaging. 2010;9:1–20. [PMC free article] [PubMed] [Google Scholar]
- [6].Walrand S, Flux G, Konijnenberg M, Valkema R, Krenning E, Lhommel R, et al. Dosimetry of yttrium-labelled radiopharmaceuticals for internal therapy: 86Y or 90Y imaging? Eur J Nucl Med Mol Imaging. 2011;38:57–68. doi: 10.1007/s00259-011-1771-7. [DOI] [PubMed] [Google Scholar]
- [7].Pauwels S, Barone R, Walrand S, Borson-Chazot F, Valkema R, Kvols LK, et al. Practical dosimetry of peptide receptor radionuclide therapy with 90Y-labeled Somatostatin analogs. J Nucl Med. 2005;46:92S–8S. [PubMed] [Google Scholar]
- [8].Nayak TK, Brechbiel MW. (86)Y based PET radiopharmaceuticals: radiochemistry and biological applications. Med Chem. 2011;7:380–8. doi: 10.2174/157340611796799249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Price EW, Orvig C. Matching chelators to radiometals for radiopharmaceuticals. Chem Soc Rev. 2014;43:260–90. doi: 10.1039/c3cs60304k. [DOI] [PubMed] [Google Scholar]
- [10].Banerjee SR, Foss CA, Pullambhatla M, Wang Y, Srinivasan S, Hobbs R, et al. Preclinical evaluation of 86Y-labeled inhibitors of prostate-specific membrane antigen for dosimetry estimates. J Nucl Med. 2015 doi: 10.2967/jnumed.114.149062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Jødal L, Loirec CL, Champion C. Positron range in PET imaging: non-conventional isotopes. Phys Med Biol. 2014;59:7419. doi: 10.1088/0031-9155/59/23/7419. [DOI] [PubMed] [Google Scholar]
- [12].Lubberink M, Herzog H. Quantitative imaging of 124I and 86Y with PET. Eur J Nucl Med Mol Imaging. 2011;38:10–8. doi: 10.1007/s00259-011-1768-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Nayak TK, Garmestani K, Milenic DE, Baidoo KE, Brechbiel MW. HER1-targeted 86Y-Panitumumab possesses superior targeting characteristics than 86Y-Cetuximab for PET imaging of human malignant mesothelioma tumors xenografts. PLoS One. 2011;6:e18198. doi: 10.1371/journal.pone.0018198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Nayak TK, Garmestani K, Baidoo KE, Milenic DE, Brechbiel MW. PET imaging of tumor angiogenesis in mice with VEGF-A–targeted 86Y-CHX-A″-DTPA-bevacizumab. Int J Cancer. 2011;128:920–6. doi: 10.1002/ijc.25409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Nayak T, Regino C, Wong K, Milenic D, Garmestani K, Baidoo K, et al. PET imaging of HER1-expressing xenografts in mice with 86Y-CHX-A″-DTPA-cetuximab. Eur J Nucl Med Mol Imaging. 2010;37:1368–76. doi: 10.1007/s00259-009-1370-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Nayak TK, Garmestani K, Baidoo KE, Milenic DE, Brechbiel MW. PET imaging of tumor angiogenesis in mice with VEGF-A–targeted 86Y-CHX-A″-DTPA-bevacizumab. Int J Cancer. 2010;128:920–6. doi: 10.1002/ijc.25409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Milenic DE, Wong KJ, Baidoo KE, Nayak TK, Regino CAS, Garmestani K, et al. Targeting HER2: a report on the in vitro and in vivo pre-clinical data supporting trastuzumab as a radioimmunoconjugate for clinical trials. MAbs. 2010;2:550–64. doi: 10.4161/mabs.2.5.13054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Schneider DW, Heitner T, Alicke B, Light DR, McLean K, Satozawa N, et al. In vivo Biodistribution, PET imaging, and tumor accumulation of 86Y- and 111In-Antimindin/RG-1, engineered antibody fragments in LNCaP tumor-bearing nude mice. J Nucl Med. 2009;50:435–43. doi: 10.2967/jnumed.108.055608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Wei L, Zhang X, Gallazzi F, Miao Y, Jin X, Brechbiel MW, et al. Melanoma imaging using 111In-, 86Y- and 68Ga-labeled CHX-A″-Re(Arg11)CCMSH. Nucl Med Biol. 2009;36:345–54. doi: 10.1016/j.nucmedbio.2009.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].McQuade P, Miao Y, Yoo J, Quinn TP, Welch MJ, Lewis JS. Imaging of melanoma using 64Cu- and 86Y-DOTA-ReCCMSH(Arg11), a cyclized peptide analogue of α-MSH. J Med Chem. 2005;48:2985–92. doi: 10.1021/jm0490282. [DOI] [PubMed] [Google Scholar]
- [21].Palm S, Enmon RM, Matei C, Kolbert KS, Xu S, Zanzonico PB, et al. Pharmacokinetics and Biodistribution of 86Y-trastuzumab for 90Y dosimetry in an ovarian carcinoma model: correlative MicroPET and MRI. J Nucl Med. 2003;44:1148–55. [PubMed] [Google Scholar]
- [22].Rösch F, Herzog H, Stolz B, Brockmann J, Köhle M, Mühlensiepen H, et al. Uptake kinetics of the somatostatin receptor ligand [86Y]DOTA-d Phe1-Tyr3-octreotide ([86Y]SMT487) using positron emission tomography in non-human primates and calculation of radiation doses of the 90Y-labelled analogue. Eur J Nucl Med Mol Imaging. 1999;26:358–66. doi: 10.1007/s002590050398. [DOI] [PubMed] [Google Scholar]
- [23].Zeglis B, Holland J, Lebedev A, Cantorias M, Lewis J. Radiopharmaceuticals for imaging in oncology with special emphasis on positron-emitting agents. In: Strauss HW, Mariani G, Volterrani D, Larson SM, editors. Nuclear oncology. New York: Springer: 2013. pp. 35–78. [Google Scholar]
- [24].Zeglis BM, Lewis JS. A practical guide to the construction of radiometallated bioconjugates for positron emission tomography. Dalton Trans. 2011;40:6168–95. doi: 10.1039/c0dt01595d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Ramogida CF, Orvig C. Tumour targeting with radiometals for diagnosis and therapy. Chem Commun. 2013;49:4720–39. doi: 10.1039/c3cc41554f. [DOI] [PubMed] [Google Scholar]
- [26].Price EW, Cawthray JF, Adam MJ, Orvig C. Modular syntheses of H4octapa and H2dedpa, and yttrium coordination chemistry relevant to 86Y/90Y radiopharmaceuticals. Dalton Trans. 2014;43:7176–90. doi: 10.1039/c4dt00239c. [DOI] [PubMed] [Google Scholar]
- [27].Price EW, Zeglis BM, Cawthray JF, Lewis JS, Adam MJ, Orvig C. What a difference a carbon makes: H4octapa vs. H4C3octapa, ligands for In-111 and Lu-177 radiochemistry. Inorg Chem. 2014;53:10412–31. doi: 10.1021/ic501466z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Price EW, Zeglis BM, Cawthray JF, Ramogida CF, Ramos N, Lewis JS, et al. H4octapa-trastuzumab: versatile acyclic chelate system for 111In and 177Lu imaging and therapy. J Am Chem Soc. 2013;135:12707–21. doi: 10.1021/ja4049493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Price EW, Cawthray JF, Bailey GA, Ferreira CL, Boros E, Adam MJ, et al. H4octapa: an acyclic Chelator for 111In radiopharmaceuticals. J Am Chem Soc. 2012;134:8670–83. doi: 10.1021/ja3024725. [DOI] [PubMed] [Google Scholar]
- [30].Camera L, Kinuya S, Garmestani K, Wu C, Brechbiel MW, Pai LH, 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]
- [31].Harrison A, Walker CA, Parker D, Jankowski KJ, Cox JPL, Craig AS, 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]
- [32].Chong H-s, Garmestani K, Ma D, Milenic DE, Overstreet T, Brechbiel MW. Synthesis and biological evaluation of novel macrocyclic ligands with pendent donor groups as potential yttrium chelators for radioimmunotherapy with improved complex formation kinetics. J Med Chem. 2002;45:3458–64. doi: 10.1021/jm0200759. [DOI] [PubMed] [Google Scholar]
- [33].Chong H-S, Song HA, Ma X, Milenic DE, Brady ED, Lim S, et al. Novel bimodal bifunctional ligands for radioimmunotherapy and targeted MRI. Bioconjug Chem. 2008;19:1439–47. doi: 10.1021/bc800050x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Kang CS, Sun X, Jia F, Song HA, Chen Y, Lewis M, et al. Synthesis and preclinical evaluation of bifunctional ligands for improved chelation chemistry of 90Y and 177Lu for targeted radioimmunotherapy. Bioconjug Chem. 2012;23:1775–82. doi: 10.1021/bc200696b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Holland JP, Sheh Y, Lewis JS. Standardized methods for the production of high specific-activity zirconium-89. Nucl Med Biol. 2009;36:729–39. doi: 10.1016/j.nucmedbio.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Zeglis BM, Mohindra P, Weissmann GI, Divilov V, Hilderbrand SA, Weissleder R, et al. Modular strategy for the construction of radiometalated antibodies for positron emission tomography based on inverse electron demand Diels–Alder click chemistry. Bioconjug Chem. 2011;22:2048–59. doi: 10.1021/bc200288d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Holland JP, Divilov V, Bander NH, Smith-Jones PM, Larson SM, Lewis JS. 89Zr-DFO-J591 for immunoPET of prostate-specific membrane antigen expression in vivo. J Nucl Med. 2010;51:1293–300. doi: 10.2967/jnumed.110.076174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Carlin S, Zhang H, Reese M, Ramos NN, Chen Q, Ricketts S-A. A comparison of the imaging characteristics and microregional distribution of 4 hypoxia PET tracers. J Nucl Med. 2014;55:515–21. doi: 10.2967/jnumed.113.126615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].DeFazio-Eli L, Strommen K, Dao-Pick T, Parry G, Goodman L, Winslow J. Quantitative assays for the measurement of HER1–HER2 heterodimerization and phosphorylation in cell lines and breast tumors: applications for diagnostics and targeted drug mechanism of action. Breast Cancer Res. 2011;13:R44. doi: 10.1186/bcr2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.