Abstract
This study sought to evaluate [99mTc(HYNIC-Galacto-RGD2)(tricine)(TPPTS)] (99mTc-Galacto-RGD2: HYNIC = 6-hydrazinonicotinyl; Galacto-RGD2 = Glu[cyclo[Arg-Gly-Asp-D-Phe-Lys(SAA-PEG2-(1,2,3-triazole)-1-yl-4-methylamide)]]2 (SAA = 7-amino-L-glycero-L-galacto-2,6-anhydro-7-deoxy-heptanamide, and PEG2 = 3,6-dioxaoctanoic acid); and TPPTS = trisodium triphenylphosphine-3,3',3"-trisulfonate) as a new radiotracer for tumor imaging. Galacto-RGD2 was prepared via the copper (I)-catalyzed 1,3-dipolar azide–alkyne Huisgen cycloaddition. HYNIC-Galacto-RGD2 was prepared by reacting Galacto-RGD2 with sodium succinimidyl 6-(2-(2-sulfonatobenzaldehyde)hydrazono)nicotinate (HYNIC-OSu) in the presence of diisopropylethylamine, and was evaluated for its integrin αvβ3 binding affinity against 125I-echistatin bound to U87MG glioma cells. The IC50 value for HYNIC-Galacto-RGD2 was determined to be 20 ± 2 nM. 99mTc-Galacto-RGD2 was prepared in high specific activity (~185 GBq/µmol) and high radiochemical purity (>95%), and was evaluated in athymic nude mice bearing U87MG glioma xenografts for its tumor-targeting capability and biodistribution. The tumor uptake of 99mTc-Galacto-RGD2 was 10.30 ± 1.67, 8.37 ± 2.13, 6.86 ± 1.33 and 5.61 ± 1.52 %ID/g at 5, 30, 60 and 120 min p.i., respectively, which was in agreement with high integrin αvβ3 expression on glioma cells and neovasculature. Its lower uptake in intestines, lungs and spleen suggests that 99mTc-Galacto-RGD2 has advantages over 99mTc-3P–RGD2 ([99mTc(HYNIC-3P–RGD2)(tricine)(TPPTS)]: 3P–RGD2 = PEG4-E[PEG4-c(RGDfK)]2; PEG4 = 15-amino-4,7,10,13-tetraoxapentadecanoic acid) for imaging tumors in the chest and abdominal regions. U87MG tumors were readily detected by SPECT and the tumor uptake of 99mTc-Galacto-RGD2 was integrin αvβ3-specific. 99mTc-Galacto-RGD2 also had very high metabolic stability. On the basis of results from this study, it was concluded that 99mTc-Galacto-RGD2 is an excellent radiotracer for imaging integrin αvβ3-positive tumors and related metastases.
Keywords: integrin αvβ3, 99mTc-labeling, RGD-containing glycopeptide, tumor imaging, SPECT
INTRODUCTION
Integrin αvβ3 plays a significant role in tumor angiogenesis and metastasis.1–8 It is a receptor for the extracellular matrix proteins (e.g.vitronectin, fibronectin, fibrinogen, lamin, collagen, Von Willibrand’s factor and osteoponin) with the exposed arginine-glycine-aspartic (RGD) peptide sequence.9–11 Integrin αvβ3 is expressed at low levels on epithelial cells and mature endothelial cells, but it is overexpressed on activated endothelial cells of neovasculature and some tumor cells. Thus, integrin αvβ3 is considered an interesting molecular target for early cancer detection.12–18
Cyclic RGD peptides, such as E[c(RGDfK)]2 (Figure 1: RGD2) and E{E[c(RGDfK)]2}2 (RGD4), are potent integrin αvβ3 receptor antagonists.10,11,16,18 Over the last several years, many radiolabeled (99mTc, 18F, 64Cu, 68Ga and 111In) multimeric cyclic RGD peptides have been evaluated as integrin αvβ3-targeted radiotracers for tumor imaging by single photon emission computed tomography (SPECT) or positron emission tomography (PET).19–39 It was shown that these cyclic RGD peptides were able to bind integrin αvβ3 with high specificity.16,18 Multiple RGD moieties were utilized to maximize their integrin αvβ3 binding affinity and the radiotracer tumor uptake.15,16,18 It was found that radiolabeled multimeric RGD peptides had significantly higher tumor uptake with much longer tumor retention time than their monomeric counterparts.16,18,24–39 It was also found that cyclic RGD peptide tetramers, such as RGD4, are bivalent in binding to integrin αvβ3 even though they have four identical RGD motifs.36,38
Figure 1.
The structure of cyclic RGD peptides (c(RGDfK), RGD2, 3P–RGD2, and Galacto-RGD2) and their corresponding ternary ligand complexes [99mTc(HYNIC-BM)(tricine)(TPPTS)] (BM = biomolecule; 99mTc-RGD2: BM = RGD2; 99mTc-3P–RGD2: BM = 3P–RGD299mTc-Galacto-RGD2: BM = Galacto-RGD2. The 7-amino-L-glycero-L-galacto-2,6-anhydro-7-deoxyheptanamide (SAA) and 1,2,3-triazole moieties are used to increase the hydrophilicity of 99mTc-Galacto-RGD2, and minimize its accumulation in normal organs, such as intestines, lungs and spleen.
We have been interested in radiolabeled cyclic RGD peptides as PET or SPECT radiotracers for molecular imaging of solid tumors.30–44 [99mTc(HYNIC-3P–RGD2)(tricine)(TPPTS)] (Figure 1: 99mTc-3P–RGD2: HYNIC = 6-hydrazinonicotinyl; 3P–RGD2 = PEG4-E[PEG4-c(RGDfK)]2; PEG4 = 15-amino-4,7,10,13-tetraoxapentadecanoic acid; and TPPTS = trisodium triphenylphosphine-3,3',3"-trisulfonate) is a 99mTc-labeled cyclic RGD peptide dimer.31 It could be prepared from 99mTcO4‒ with high specific activity using a kit formulation.31,40.99mTc-3P–RGD2 showed very high tumor uptake and rapid renal clearance in glioma-bearing nude mice,31 and its tumor uptake correlated well linearly with the integrin αvβ3 expression levels in five different tumor-bearing small animal models.42. 99mTc-3P–RGD2 SPECT/CT has been successfully used to quantify the tumor uptake and for noninvasive monitoring of progression of breast cancer lung metastases.43,44 It is currently under clinical investigation as a new radiotracer for tumor imaging in cancer patients.45,46
However, 99mTc-3P–RGD2 has high uptake in intestines and spleen,31,45,46 which makes it difficult to image tumors in the abdominal region. To minimize excess radioactivity accumulation in normal organs, we designed and prepared a new cyclic RGD peptide dimer, Galacto-RGD2 (Figure 1), in which the 7-amino-L-glycero-L-galacto-2,6-anhydro-7-deoxyheptanamide (SAA) groups were used to bridge the two cyclic RGD moieties. SAA has been successfully used to improve pharmacokinetics of 18F-labeled cyclic RGD peptides.19–21,47 The 1,2,3-triazole moiety was reported to have beneficial effect on pharmacokinetics of radiotracers by increasing their washout efficiency from liver and GI tract.48 With this in mind, we decided to combine the SAA and 1,2,3-triazole moieties in Galacto-RGD2. Our hypothesis is that the SAA and 1,2,3-triazole moieties will help to reduce its accumulation in normal organs, such as the intestines, liver, lungs and spleen.
In this report, we present the synthesis of Galacto-RGD2, and evaluations of its 99mTc complex [99mTc(HYNIC-Galacto-RGD2)(tricine)(TPPTS)] (Figure 1: 99mTc-Galacto-RGD2) as a new radiotracer for tumor imaging. Integrin αvβ3 binding affinity of HYNIC-Galacto-RGD2 was determined in an in vitro displacement assay. Biodistribution properties of 99mTc-Galacto-RGD2 were evaluated in the athymic nude mice bearing U87MG glioma xenografts, and compared to those of 99mTc-3P–RGD2. The main objective of this study was to demonstrate that SAA and 1,2,3-triazole moieties are indeed able to improve excretion kinetics of 99mTc radiotracer from non-cancerous organs.
EXPERIMENTAL SECTION
Materials and Instruments
Chemicals were purchased from Sigma/Aldrich (St. Louis, MO), and were used without purification unless specified. The Boc-protected glutamic acid was purchased from Senn Chemicals (San Diego, CA), and 8-azido-3,6-dioxaoctanoic acid (N3-PEG2-OH) was from Peptides International Inc. (Louisville, KY). The protected peptide [cyclo[Arg(Pbf)-Gly-Asp(OtBu)-D-Phe-Lys(SAA)]] (Galacto-RGD) was prepared using the procedure reported in the literature.47 Sodium succinimidyl 6-(2-(2-sulfonatobenzaldehyde)hydrazono)nicotinate (HYNIC-OSu) was prepared according to the literature method.49 [99mTc(HYNIC-3P–RGD2)(tricine)(TPPTS)] (99mTc-3P–RGD2) was prepared using the procedure described in our previous reports.31,40 Na99mTcO4 was obtained from Cardinal HealthCare® (Chicago, IL). The MALDI (matrix-assisted laser desorption ionization) mass spectral data were collected on an Applied Biosystems Voyager DE PRO mass spectrometer (Framingham, MA), the Department of Chemistry, Purdue University. Melting point was determined on a Buchi B-540 melting point apparatus and was uncorrected. Optical rotation was determined on a DIP-370 polarimeter (Jasco, Easton, MD). Thin layer chromatography was performed using Merck F254 silica gel plates (EMD, Darmstadt, Germany). NMR spectra were recorded with a Varian Unity 500 Plus spectrometer (Palo Alto, CA) in DMSO-d6.
HPLC Methods
The RP-HPLC (Method 1) used a Shimadzu preparative system consisting of two LC-8A pumps controlled by a Shimadzu SCL-10A system, and a Shimadzu UV/VIS detector SPD-6A (λ=220 nm) equipped with a preparative flow cell, a Prochrom preparative HPLC column (50 mm × 35 cm,100 Å pore size, Phenomenex, Torrance, CA). The flow rate was 100 mL/min with a gradient phase starting from 90% A (0.05 % TFA in H2O) and 10 % B (0.05 % TFA in CH3CN) at 0 min to 60%A and 40 % B at 60 min. HPLC Method 2 used a LabAlliance HPLC system (Scientific Systems, Inc., State College, PA) equipped with a UV/vis detector (λ=254 nm) and Zorbax C18 column (9.4 mm × 250 mm, 100 Å pore size; Agilent Technologies, Santa Clara, CA). The flow rate was 2.5 mL/min with a mobile phase being 90% A and 10% B at 0 min to 80%A and 20% B at 5 min, and to 70%A and 30% B at 20 min. The radio-HPLC (Method 3) used the LabAlliance HPLC system equipped with a β-ram IN/US detector (Tampa, FL) and Zorbax C18 column (4.6 mm × 250 mm, 300 Å pore size; Agilent Technologies, Santa Clara, CA). The flow rate was 1 mL/min. The mobile phase was isocratic for the first 5 min with 90% A (25 mM NH4OAc, pH = 6.8) and 10% B (acetonitrile), followed by a gradient mobile phase going from 90% A and 10% B at 5 min to 60% A and 40% B at 20 min.
Boc-L-Glutamic Acid Bis-Propargyl Amide (1)
To a solution of Boc-glutamic acid (2.47 g, 10 mmol) and propargylamine hydrochloride (2.11 g, 23 mmol) in DMF (30 mL) were added diphenylphosphoryl azide (DPPA: 5.2 mL, 24 mmol) and triethylamine (Et3N, 3.2 mL, 23 mmol) at 0 °C under vigorous stirring. The reaction was allowed to proceed overnight at 0 °C. The pH value of the reaction mixture was kept at 7 ~ 8 with Et3N. The reaction mixture was taken into EtOAc (500 mL) and washed with 0.2 N HCl (2 × 200 mL), brine (100 mL), saturated NaHCO3 solution (3 × 200 mL), and brine (2 × 100 mL). After drying over Na2SO4, EtOAc was removed to yield 4.16 g of crude product, which upon trituration with hot isopropyl ether afforded a white solid (2.66 g, 82.9 %). Rf (EtOAc/Hexane/AcOH 2:1:0.01) = 0.20, melting point 146.2–146.5°C, [α] D25 10.4° (c = 1, CHCl3). ESI-MS: m/z = 344.2 [M + Na]+; 322.2 [M + H]+. 1H NMR (DMSO-d6, chemical shift δ in ppm, and atom labels according to the structure below): δ 8.25 [1H, bt, J=5.6 Hz, N(1a)-H], 8.24 [1H, bt, J=5.6 Hz, N(5a)-H], 6.88 [1H, d, J= 8.0 Hz, N(2a)-H], 3.87 [1H, m, C(2)-H], 3.85 [2H, dd, J=5.6 Hz, J=2.3 Hz, C(1b)-H2], 3.82 [2H, dd, J=5.6 Hz, J=2.6 Hz, C(5b)-H2], 3.10 [1H, t, J=2.3 Hz, C(1d)-H], 3.08 [1H, t, J=2.6 Hz, C(5d)-H], 2.10 [2H, m, C(4)-H2], 1.82 [1H, m, C(3)- H2], 1.67 [1H, m, C(3)- H2], and 1.37 [9H, s, C(2e)-H3]. 13C NMR (DMSO-d6, chemical shift δ in ppm): 171.5 (C-1), 171.2 (C-5), 155.2 (C-2b), 81.2 (C-1c), 81.1 (C-5c), 78.1 (C-2d), 73.0 (C-5d), 72.9 (C-1d), 53.8 (C-2), 31.6 (C-4), 28.2 (C-2e), 27.9 (C-1b), 27.8 (C3 and C-5b). Anal. Calcd for C16H23N3O4: C, 59.80; H, 7.21; N, 13.08. Found: C, 59.78; H, 7.19; N, 13.00.
Cyclo[Arg(Pbf)-Gly-Asp(OtBu)-D-Phe-Lys(SAA-PEG2-N3)] (2)
To a solution of cyclo[Arg(Pbf)-Gly-Asp(OtBu)-D-Phe-Lys(SAA)] (1.10 g, 1.0 mmol) and N3-PEG2-OH (0.28 g, 1.48 mmol) in DMF (15 mL) was added DPPA (0.43 mL, 2.0 mmol) at 0 °C with stirring, followed by the addition of Et3N (0.21 mL, 1.51 mmol). The reaction mixture was kept at 0 °C for 25 h, with occasional adjustments of the pH (7 – 8), with Et3N. After the reaction, the mixture was diluted with 0.4 N HCl (100 mL) and extracted with EtOAc (3 × 300 mL). The combined organic extracts were concentrated. The precipitated product was collected by filtration, washed with cold Et2O, and dried in vacuo to afford 0.81 g of 2 in 63.7 % yield as a colorless solid. ESI-MS: m/z = 945.55 [M + H]+ (M = 945.43 calcd. for [C40H59N13O14]).
Boc-Glu[cyclo[Arg(Pbf)-Gly-Asp(OtBu)-D-Phe-Lys(SAA-PEG2-(1,2,3-triazole)-1-yl-4-methylamide)]]2 (3)
To a stirred solution of 1 (94 mg, 0.29 mmol) and 2 (750 mg, 0.59 mmol) in a mixture of H2O (2.9 mL) and t-BuOH (5.8 mL) were added 1.73 mL of 0.2 M CuSO4 5H2O and 1.73 mL of 0.5 M sodium ascorbate solution at room temperature. The mixture was stirred for 70 min, then diluted with 60 mL 1-BuOH and 40 mL H2O. The organic layer was washed with H2O (2 × 30 mL) and concentrated under reduced pressure. Addition of Et2O resulted in a precipitate, which was collected by filtration, washed with Et2O, and dried to give 760 mg of 3. The yield was 91.5 %. ESI-MS: m/z = 2249.05 [M+H]+ (M = 2249.05 calcd. for [C96H145N29O34]).
H-Glu[cyclo[Arg-Gly-Asp-D-Phe-Lys(SAA-PEG2-(1,2,3-triazole)-1-yl-4-methylamide)]]2 (Galacto-RGD2)
The protected Galacto-RGD2 (750 mg, 0.26 mmol) was dissolved in 20 mL of a solution TFA/TIPS/H2O (95/2.5/2.5, v/v/v) at room temperature. After 2 h, the reaction mixture was concentrated under reduced pressure. Crude Galacto-RGD2 was precipitated with cold Et2O, collected by centrifugation, washed with Et2O (3 × 30 mL), and dried in vacuo to give 690 mg of white solid, which was purified by RP-HPLC (Method 1) to yield Galacto-RGD2 (105 mg, 16.2 %) as a TFA salt. The HPLC purity was 99.3 %. ESI-MS: m/z = 2149.12 [M+H]+ (M = 2149.05 calcd. for [C91H137N29O34]).
HYNIC-Glu[cyclo[Arg-Gly-Asp-D-Phe-Lys(SAA-PEG2-(1,2,3-triazole)-1-yl-4-methylamide)]]2 (HYNIC-Galacto-RGD2)
HYNIC-OSu (5.4 mg, 12 µmol) and Galacto-RGD2 (6.5 mg, 3 µmol) were dissolved in DMF (2 mL). After addition of excess diisopropylethylamine (DIEA, 50 µmol), the reaction mixture was stirred for ~10 days at room temperature. To the reaction mixture was added 2 mL of water after completion of conjugation. The pH value was then adjusted to 3 – 4 using TFA. The product was separated from the reaction mixture by HPLC (Method 2). Fractions at 14.5 min were collected. Lyophilization of collected fractions afforded HYNIC-Galacto-RGD2. The yield was 3.5 mg (~48%). MALDI-MS: m/z = 2452.3 for [M + H]+ (M = 2451.03 calcd. for [C104H146N32O36S]).
99mTc-Labeling
To a lyophilized vial containing 20 µg HYNIC-Galacto-RGD2, 7 mg TPPTS, 6.5 mg tricine, 40 mg mannitol, 38.5 mg disodium succinate hexahydrate and 12.7 mg succinic acid was added 1.0 – 1.5 mL of Na99mTcO4 solution (~1,110 MBq in saline). The reconstituted vial was then heated in a boiling water bath for ~30 min. After radiolabeling, a sample of the resulting solution was analyzed by radio-HPLC (Method 3). The radiochemical purity was >90% for imaging and biodistribution studies. The solution stability was monitored by radio-HPLC for 24 h.
Dose Preparation
For biodistribution, doses were prepared by dissolving 99mTc-Galacto-RGD2 in saline to a concentration of ~1 MBq/mL. For SPECT/CT imaging, doses were prepared by dissolving 99mTc-Galacto-RGD2 in saline to ~370 MBq/mL. In blocking experiment, RGD2 was dissolved in the dose solution to 3.5 mg/mL. The resulting dose solution was filtered with a 0.20 µm Millex-LG filter before being injected into animals. Each animal was injected with ~0.1 mL of the dose solution.
Tumor Cell Culture
The U87MG cell line was obtained from ATCC (American Type Culture Collection, Manassas, VA). U87MG cells were cultured in the Minimum Essential Medium, Eagle with Earle’s Balanced Salt Solution (non-essential amino acids sodium pyruvate), and were supplemented with 10% fetal bovine serum (FBS, ATCC) and 1% penicillin and streptomycin solution at 37 °C in a humidified atmosphere of 5% CO2 in air. Cells were grown as monolayers and were harvested or split when they reached 90% confluence to maintain exponential growth.
In Vitro Whole-Cell Integrin αvβ3 Binding Assay
The integrin binding affinity of cyclic RGD peptides were assessed via a cellular competitive displacement assay using125 I-echistatin (Perkin Elmer, Branford, CT) as the integrin-specific radioligand.31,32 Briefly, the filter multiscreen DV plates (Millipore, Billerica, MA) were seeded with 1×105 U87MG cells in binding buffer (20 mM Tris, 150 mM NaCl, 2 mM CaCl2, 1 mM MnCl2, 1 mM MgCl2, 0.1% (wt/vol) bovine serum albumin; and pH 7.4) and 125I-echistatin (0.75 – 1.0 kBq) in the presence of increasing concentrations of the cyclic RGD peptide, incubated for 2 h at room temperature. After removing unbound 125I-echistatin, the hydrophilic PVDF filters were washed 3× with the binding buffer, and then collected. Radioactivity was determined using a Perkin Elmer Wizard – 1480 γ-counter (Shelton, CT). Experiments were carried out twice with triplicates. IC50 values were calculated by fitting experimental data with nonlinear regression using GraphPad Prism™ (GraphPad Software, Inc., San Diego, CA), and were reported as an average plus/minus standard deviation.
Animal Models
Biodistribution and imaging studies were performed in compliance with the NIH animal experimentation guidelines (Principles of Laboratory Animal Care, NIH Publication No. 86–23, revised 1985). The protocol was approved by the Purdue University Animal Care and Use Committee (PACUC). Female athymic nu/nu mice (4 – 5 weeks) were purchased from Harlan (Indianapolis, IN), and were inoculated subcutaneously with 5 × 106 of U87MG cells into the shoulder flank of each animal. Four weeks after inoculation, the tumor size was 0.1 – 0.5 g, and animals were used for biodistribution and imaging studies.
Biodistribution Protocol
The tumor-bearing mice (20 – 25 g) were randomly selected, and each animal was administered with ~0.1 MBq of 99mTc-Galacto-RGD2 (~2 ng HYNIC-Galacto-RGD2) by tail vein injection. Animals (5 – 8) were sacrificed by sodium pentobarbital overdose (~200 mg/kg) at 5, 30, 60 and 120 min post-injection (p.i.). Blood, tumors and normal organs (brain, eyes, heart, spleen, lungs, liver, kidneys, muscle and intestine) were harvested, washed with saline, dried with absorbent tissue, weighed, and counted on a Perkin Elmer Wizard – 1480 γ-counter. The organ uptake was calculated as the percentage of injected dose per gram of wet tissue (%ID/g). The blocking experiment was performed using RGD2 as the blocking agent in six animals. Each animal was administered with ~0.1 MBq of 99mTc-Galacto-RGD2 along with ~350 µg (~14 mg/kg) of RGD2. Biodistribution data (%ID/g) and tumor-to-background (T/B) ratios were expressed as the average plus standard deviation.
SPECT/CT Imaging
SPECT/CT images were obtained using a u-SPECT-II/CT scanner (Milabs, Utrecht, The Netherlands) equipped with a 0.6 mm multi-pinhole collimator. The glioma-bearing mouse was injected with ~37 MBq of 99mTc-Galacto-RGD2 (~0.8 µg HYNIC-Galacto-RGD2) in 0.1 mL saline via the tail vein. At 60 min p.i., the animal was placed into a shielded chamber connected to an isoflurane anesthesia unit (Univentor, Zejtun, Malta). Anesthesia was induced using an air flow rate of 350 mL/min and ~3.0% isoflurane. After induction of anesthesia, the animal was immediately placed supine on the scanning bed. The air flow rate was then reduced to ~250 mL/min with ~2.0% isoflurane. Rectangular scan in the regions of interest (ROIs) from SPECT and CT were selected on the basis of orthogonal optical images provided by the integrated webcams. After SPECT acquisition (75 projections over 30 min per frame, 2 frames), the animal was then transferred into the attached CT scanner and imaged using the ‘normal’ acquisition settings (2 degree intervals) at 45 kV and 500 µA. After CT acquisition, the animal was allowed to recover in a lead-shielded cage.
Image Reconstruction and Data Processing
SPECT reconstruction was performed using a POSEM (pixelated ordered subsets by expectation maximization) algorithm with 6 iterations and 16 subsets. CT data were reconstructed using a cone-beam filtered back-projection algorithm (NRecon v1.6.3, Skyscan). After reconstruction, the SPECT and CT data were automatically co-registered according to the movement of the robotic stage, and then re-sampled to equivalent voxel sizes. Co-registered images were further rendered and visualized using the PMOD software (PMOD Technologies, Zurich, Switzerland). A 3D–Guassian filter (0.8 mm FWHM) was applied to smooth noise, and the LUTs (look up tables) were adjusted for good visual contrast. The reconstructed images were visualized as both orthogonal slices and maximum intensity projections.
Radioactivity Quantification
Radiation sources of known amount of radioactivity were imaged and reconstructed using the same scanning protocol described above. A standard curve was generated to correlate the pixel intensities in the reconstructed images to the radioactivity as measured by a γ-counter. The tumor delineation was performed on CT and SPECT images according to the literature method.43,44 Briefly, the ROIs (regions of interest) were drawn manually to cover the entire tumor based on transverse view of the CT image. For the tumor delineation with SPECT, a threshold of 50% or more of the maximum pixel value on the SPECT image was chosen. In order to accurately represent the entire tumor, SPECT/CT was also employed to delineate the tumor ROIs. The tumor volume and radioactivity counts were generated by using the PMOD software (PMOD Technologies, Zurich, Switzerland). The amount of radioactivity in each tumor was calculated according to the above mentioned standard curve. The tumor uptake of 99mTc-Galacto-RGD2 was expressed the percentage of injected dose per unit volume (%ID/cm3), and was compared with the data from biodistribution in the same tumor-bearing animal model.
Metabolism
Athymic nude mice (n = 3) without tumors were used for metabolism study. Each animal was administered with ~10 MBq of 99mTc-Galacto-RGD2 via the tail vein. The urine samples were collected at 30 min and 120 min p.i. by manual void, and were mixed with equal volume of 50% acetonitrile aqueous solution. The mixture was centrifuged at 8,000 rpm. The supernatant was collected and filtered with a 0.20 µm Millex-LG filter to remove foreign particles. The filtrate was analyzed by HPLC (Method 2). Feces samples were collected at 120 min p.i. and suspended in 20% acetonitrile aqueous solution after homogenization. The resulting mixture was vortexed for ~5 min. After centrifuging at 8,000 rpm, the supernatant was collected and filtered with a 0.20 µm Millex-LG filter to remove foreign particles. The filtrate was then analyzed by HPLC (Method 2). The percentage of radioactivity recovery was >95% (by γ-counting) for both urine and feces samples.
Cultured Primary Tumor Cells
The xenografted U87MG tumors were harvested, and were immersed in a growth medium immediately after gamma counting. Tumor tissues were rinsed twice with the Hanks’ solution and dissected into small pieces with razor blades. Small tumor pieces were digested with 0.25% Trypsin (1 mM EDTA, without Ca2+ and Mg2+) solution at 37 °C for 30 min, followed by vigorous pipetting and filtration through a 40 µm mesh nylon screen. After centrifugation at 1000 rpm for 5 min, the pellet was re-suspended in the culture media and incubated at 37 °C in a humidified atmosphere containing 5% CO2. The purity of tumor cells could reach ~90% after two passages, and the third passage was used for cellular immunostaining studies.
Cellular Immunostaining
The primary U87MG glioma cells were seeded into 8-well chamber slides, and allowed to spread for >24 h. Cells were fixed in −20 °C methanol for 5 min and rinsed with PBS. Cells were incubated in 5% BSA for 30 min to block nonspecific binding, and then added Alexa Fluor 488 conjugated mouse anti-human integrin αvβ3 monoclonal antibody LM609 (1:100, Millipore, Billerica, MA) for 1 h at room temperature, or incubated with rabbit anti-human and murine β3 antibody followed by incubation with Alexa Fluor 594 conjugated goat anti-rabbit IgG (Santa Cruz, CA). After washing with PBS buffer, the slides were mounted with Dapi-Fluoromount-G (SouthernBiotech, Birmingham, AL). The fluorescence was visualized with an Olympic BX51 microscope (Olympus America Inc., Center Valley, PA).
Tumor Tissue Immunohistochemistry
The U87MG tumors harvested from tumor-bearing mice were immediately snap-frozen in the OCT (optical cutting temperature) solution, and were then cut into slices (5 µm). After thorough drying at room temperature, slices were fixed with ice-cold acetone for 10 min, and dried in the air for 20 min. Sections were blocked with 10% goat serum for 30 min at room temperature, and then were incubated with the hamster anti-integrin β3 antibody (1:100, BD Biosciences, San Jose, CA) and rat anti-CD31 antibody (1:100, BD Biosciences) for 1 h at room temperature. After incubating with the Cy3-conjugated goat anti-hamster and fluorescein isothiocyanate (FITC)-conjugated goat anti-rat secondary antibodies (1:100, Jackson ImmunoResearch Inc., West Grove, PA) and washing with PBS, the fluorescence was visualized with an Olympus fluorescence microscope (Olympus America Inc., Center Valley, PA). All the pictures were taken under 200x magnification with the same exposure time. Brightness and contrast adjustments were made equally to all images.
Statistical Analysis
All experimental data were expressed as the average plus/minus standard deviation. Statistical analysis was performed by one-way analysis of variance (ANOVA) followed by the Newman-Keuls test for multiple comparisons. The level of significance was set at p < 0.05.
RESULTS
Synthesis of Galacto-RGD2
Compound 1, a key intermediate for preparation of Galacto-RGD2, was prepared (Chart I: ~83% yield) from the reaction of Boc-glutamic acid with two equivalents of propargylamine in the presence of DPPA, which is a coupling agent with low risk of racemization. 50,51 DPPA was also employed in the preparation of 2. Synthesis of 3 was accomplished in >90% yield by the Cu(I)-catalyzed 1,3-dipolar azide–alkyne Huisgen cycloaddition. It was not necessary to protect the hydroxyl groups in the SAA moieties during this reaction. Deprotection of the Pbf and t-Butyl protecting groups in 3 under acidic conditions resulted in Galacto-RGD2, which was characterized by HPLC (Figure SI1), ESI-MS (Figure SI2) and NMR (Figures SI3 – SI8: DQF-COSY, NOESY, ROESY, TOCSY, HSQC, and HMBC). The ESI-MS and NMR data (Tables SI1 and SI2) were completely consistent with the structure and composition proposed for Galacto-RGD2.
Chart I.
Syntheses of Galacto-RGD2
Synthesis of HYNIC-Galacto-RGD2
HYNIC-Galacto-RGD2 was prepared from the reaction of Galacto-RGD2 with HYNIC-OSu in the presence of excess DIEA. The conjugation reaction was slow, and took about 10 days to complete at room temperature. This was likely due to the fact that the primary amine in Galacto-RGD2 is stereochemically “hindered” by two bulky Galacto-RGD monomers. HYNIC-Galacto-RGD2 was purified by semi-prep HPLC and characterized by ESI-MS. Its HPLC purity was > 95% before being used for 99mTc-labeling. ESI-MS data was consistent with the proposed composition for HYNIC-Galacto-RGD2.
Integrin αvβ3 Binding Affinity
Figure 2 shows the displacement curves of 125I-echistatin bound to U87MG human glioma cells in the presence of HYNIC-Galacto-RGD2. For comparison purpose, we also evaluated c(RGDyK), HYNIC-RGD2 and HYNIC-3P–RGD2 in the same assay. IC50 values were calculated to be 20±2, 32±1, 85±2 and 422±15 nM for HYNIC-Galacto-RGD2, HYNIC-3P–RGD2, HYNIC-RGD2 and c(RGDyK), respectively. The IC50 values of c(RGDyK) and HYNIC-RGD2 were close to those from our previous report.31,32 The integrin αvβ3 binding affinity follows the rank order of HYNIC-Galacto-RGD2 ≥ HYNIC-3P–RGD2 > HYNIC-RGD2 >> c(RGDyK) using the same competitive displacement assay.
Figure 2.
The competitive displacement curves of 125I-echistatin bound to U87MG human glioma cells in the presence of cyclic RGD peptides. HYNIC-3P–RGD2, HYNIC-RGD2 and c(RGDyK) were used for comparison purposes. IC50 values were calculated to be 20±2, 32±1, 85±2 and 422±15 nM for HYNIC-Galacto-RGD2, HYNIC-3P–RGD2, HYNIC-RGD2 and c(RGDyK), respectively.
Radiochemistry
99mTc-Galacto-RGD2 was prepared from the reaction of HYNIC-Galacto-RGD2 with 99mTcO4‒ in the presence of excess tricine and TPPTS. 99mTc-labeling was accomplished by heating the reaction mixture at 100 °C for 30 min. Figure 2 shows representative radio-HPLC chromatograms of 99mTc-Galacto-RGD2 in the kit matrix at 1 and 24 h post-labeling. Its radiochemical purity for was >95% without further chromatographic purification. The specific activity was ~185 GBq/µmol. It remained stable in the kit matrix for more than 24 h post-labeling.
Biodistribution
Table 1 lists the biodistribution data of 99mTc-Galacto-RGD2 in athymic nude mice bearing U87MG glioma xenografts. Its tumor uptake was 10.30 ± 1.67, 8.37 ± 2.13, 6.86 ± 1.33 and 5.61 ± 1.52 %ID/g at 5, 30, 60 and 120 min p.i., respectively. For comparison purposes, we also obtained the 60-min biodistribution date of 99mTc-3P–RGD2 in the same tumor-bearing animal model. As illustrate in Figure 4A, the tumor uptake of 99mTc-Galacto-RGD2 was close to that of 99mTc-3P–RGD2 at <60 min p.i. However, its 120-min tumor uptake (5.61 ± 1.52 %ID/g) that was significantly lower than that of 99mTc-3P–RGD2 (9.74 ± 3.22 %ID/g).31 Replacing PEG4 linkers in 99mTc-3P–RGD2 with a pair of SAA, 1,2,3-triazole and PEG2 moieties resulted in faster tumor washout of 99mTc-Galacto-RGD2. The liver uptake of 99mTc-Galacto-RGD2 was 4.00 ±1.10, 2.82 ± 1.25, 3.07 ± 2.14, and 1.54 ±0.43%ID/g with the tumor/liver ratios being 2.66 ± 0.51, 3.25 ± 0.85, 3.05 ± 1.69 and 4.03 ± 2.04 at 5, 30, 60, and 120 min p.i., respectively. Its liver uptake was comparable to that of 99mTc-3P–RGD2 over 2 h period.31 The main difference between 99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 was their uptake in intestines, lungs and spleen (Figure 4A). For example, the intestine uptake of 99mTc-Galacto-RGD2 (5.45 ± 1.47 %ID/g) was much lower (p<0.01) than that of 99mTc-3P–RGD2 (9.45 ± 1.31 %ID/g) at 60 min p.i.
Table 1.
Selected biodistribution data and tumor-to-background ratios of 99mTc-Galacto-RGD2 in athymic nude mice bearing U87MG human glioma xenografts. The tumor uptake was expressed as an average plus/minus the standard deviation.
| Organ | 5 min (n = 5) | 30 min (n = 5) | 60 min (n = 8) | 120 min (n = 8) |
| Blood | 1.10 ± 0.16 | 0.92 ± 0.33 | 0.56 ± 0.25 | 0.22 ± 0.03 |
| Brain | 0.41 ± 0.10 | 0.22 ± 0.06 | 0.35 ± 0.28 | 0.14 ± 0.12 |
| Eyes | 1.54 ± 0.49 | 1.20 ± 0.50 | 0.81 ± 0.39 | 0.52 ± 0.15 |
| Heart | 2.36 ± 0.29 | 1.65 ± 1.10 | 1.27 ± 0.61 | 0.56 ± 0.08 |
| Intestine | 3.78 ± 0.52 | 4.24 ± 1.54 | 5.45 ± 1.47 | 3.37 ± 1.32 |
| Kidney | 18.75 ± 2.79 | 13.13 ± 4.75 | 11.70 ± 4.06 | 6.04 ± 1.92 |
| Liver | 4.00 ± 0.10 | 2.82 ± 1.25 | 3.07 ± 2.14 | 1.54 ±0.43 |
| Lungs | 5.41 ± 0.58 | 4.24 ± 1.71 | 2.93 ± 0.94 | 1.31 ± 0.23 |
| Muscle | 2.73 ± 0.89 | 1.10 ± 0.25 | 0.88 ± 0.34 | 0.47 ± 0.12 |
| Spleen | 2.97 ± 0.43 | 2.53 ± 0.85 | 1.99 ± 0.71 | 1.46 ± 0.26 |
| Tumor | 10.30 ± 1.67 | 8.37 ± 2.13 | 6.86 ± 1.33 | 5.61 ± 1.52 |
| Tumor/Blood | 9.43 ± 1.19 | 9.65 ± 2.03 | 15.94 ± 8.85 | 26.48 ± 8.71 |
| Tumor/Kidney | 0.52 ± 0.14 | 0.61 ±0.12 | 0.79 ±0.40 | 0.89 ±0.48 |
| Tumor/Liver | 2.66 ± 0.51 | 3.25 ± 0.85 | 3.05 ± 1.69 | 4.03 ± 2.04 |
| Tumor/Lung | 1.90 ± 0.21 | 2.19 ± 0.75 | 2.73 ± 1.33 | 4.38 ± 1.34 |
| Tumor/Muscle | 3.98 ± 0.75 | 7.97 ± 2.53 | 9.40 ± 5.11 | 12.91 ± 4.95 |
Figure 4.
A: Comparison of the 60-min biodistribution data between 99mTc-Galacto-RGD2 (n = 8) and 99mTc-3P–RGD2 (n = 6) in the athymic nude mice bearing U87MG glioma xenografts to demonstrate the advantage of 99mTc-Galacto-RGD2 over 99mTc-3P–RGD2B: The selected 60-min biodistribution data of 99mTc-Galacto-RGD2 in the athymic nude mice (n = 8) bearing U87MG glioma xenografts with/without co-injection of RGD2 (350 µg/mouse or 14 mg/kg) to demonstrate its integrin αvβ3 specificity.
Integrin αvβ3 Specificity
Figure 4B shows the 60-min biodistribution data of 99mTc-Galacto-RGD2 in athymic nude mice (n = 6) bearing U87MG glioma xenografts in the absence/presence of excess RGD2 (350 µg/mouse or 14 mg/kg). Co-injection of excess RGD2 significantly blocked its tumor uptake (1.48 ± 0.42 %ID/g with RGD2 vs. 6.86 ± 1.33 %ID/g without RGD2). The normal organ uptake was also blocked by co-injection of excess RGD2. For example, the uptake of 99mTc-Galacto-RGD2 in the intestine, lungs and spleen was 5.56 ± 1.73, 2.19 ± 0.94, and 1.09 ± 0.44 %ID/g, respectively, without RGD2, while its uptake in the same organs was 1.82 ± 0.86, 1.52 ± 0.27, and 0.65 ± 0.07 %ID/g, respectively, in the presence of RGD2. This experiment clearly demonstrated that the tumor uptake of 99mTc-Galacto-RGD2 is integrin αvβ3-specific.
SPECT/CT Imaging
Figure 5A shows 3D (left) and transverse (right) views of SPECT/CT images of glioma-bearing mice administered with ~37 MBq of 99mTc-Galacto-RGD2 and 99mTc-3P–RGD2. 99mTc-3P–RGD2 was used for comparison purpose. Their tumor uptake was ~8.5 %ID/cm3 and ~7.0 %ID/cm3 , respectively, on the basis of SPECT quantification. The tumor was clearly visualized by SPECT/CT with excellent contrast with both radiotracers. As expected from biodistribution (Table 1),99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 displayed substantial GI uptake (Figure 5A). The SPECT/CT data clearly showed that both are excellent radiotracers for imaging U87MG gliomas. Figure 5B shows cellular immunostaining data. Figure 5C displays microscopic images of selected tumor slice stained with anti-integrin β3 (red color) or anti-CD31 (green color) antibody. CD31 was used as a biomarker for tumor blood vessels (mature and neovasculature), and was visualized with FITC (green). Integrin β3 was visualized with Cy3 (red). Yellow color (overlay between red β3 staining and green CD31 staining) indicates the presence of integrin αvβ3 on tumor neovasculature. It was clear that integrin αvβ3 was highly expressed on both U87MG glioma cells and neovasculature, which was in complete agreement with the high uptake of 99mTc-Galacto-RGD2 in glioma tumors (Table 1).
Figure 5.
A: The 3D and transverse views of SPECT/CT images of the athymic nude mice bearing U87MG human glioma xenografts. The animal was administered with ~37 MBq of 99mTc-3P–RGD2 or 99mTc-Galacto-RGD2. SPECT/CT study was designed to illustrate their utility for tumor imaging. B: Representative fluorescence microscopic images of the U87MG primary tumor cells, which were isolated from U87MG glioma tissue and stained with Alexa Fluor 488 conjugated mouse anti-human integrin αvβ3 monoclonal antibody LM609 (green) or with rabbit anti-human and murine integrin β3 antibody (red). Blue color indicates the presence of nuclei stained with DAPI (4’,6-diamidino-2-phenylindole). C: Selected microscopic images (Magnification: 200x) of the tumor slice stained with hamster anti-integrin β3 (red) and rat anti-CD31 (green) antibodies. Yellow color (red integrin β3 staining merged with green CD31 staining) indicates the presence of αvβ3 on tumor neovasculature.
Metabolism
Metabolism study was performed to show the in vivo stability of 99mTc-Galacto-RGD2. Figure 6 shows representative radio-HPLC chromatograms of 99mTc-Galacto-RGD2 in saline before being injected into the animal (A), in the urine at 30 min p.i. (B) and 120 min p.i. (C), and in the feces at 120 min p.i. (D). There were no metabolites detected in the urine sample over the 2 h study period for 99mTc-Galacto-RGD2. More than 90% of 99mTc-Galacto-RGD2 remained intact in the feces sample at 2 h p.i. Its metabolic stability was better than that of 99mTc-3P–RGD2, only 70% of which remained intact in the feces at 2 h p.i.31
Figure 6.
Typical radio-HPLC chromatograms for 99mTc-Galacto-RGD2 in the saline before injection, in the urine at 30 min p.i. and 120 min p.i., and in the feces at 120 min p.i. Small peaks after the main peak at ~13 min were at the noise level due to the extremely small amount of radioactivity isolated from feces sample for HPLC analysis.
DISCUSSION
In this study, we prepared a novel RGD peptide dimer, Galacto-RGD2, which contains a pair of SAA and 1,2,3-triazole groups. A similar approach was used to develop other integrin αvβ3-targeted radiotracers,52,53 such as 18F-RGD-K5, in which 1,2,3-triazole is formed via click reaction to attach the 18F-containing fragment to Galacto-RGD.52 In Galacto-RGD2, the SAA and 1,2,3-triazole moieties are parts of a linker with two extra PEG2 groups and a glutamic acid residue. Boc-Glutamic acid bis-propargyl amide was a key intermediate for synthesis of Galacto-RGD2. It may become an useful building block for click-type of dimerization/multimerization reactions of peptides and other organic molecules via the Cu(I)-catalyzed Huisgen cycloaddition. Click chemistry has proven valuable in development radiopharmaceuticals,52,54–60 including radiolabeled multimeric cyclic RGD peptides.55–60
The integrin αvβ3 binding affinity of HYNIC-Galacto-RGD2 (IC50 = 20±2 nM) was slightly higher than that of HYNIC-3P–RGD2 (IC50 = 32±1 nM), likely due to the rigidity of SAA and 1,2,3-triazole moieties in Galacto-RGD2 as compared to more flexible PEG4 groups in 3P–RGD2. It is important to note that IC50 values depends on the radioligand (125I-c(RGDyK) vs. 125I-echistatin) and tumor cell lines (U87MG vs. MDA-MB-435) used in the competitive assay. Caution should be taken when comparing IC50 values of cyclic RGD peptides with those reported in the literature.
99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 shared a similar tumor uptake (Figure 4A) despite of slightly higher integrin αvβ3 binding affinity of HYNIC-Galacto-RGD2 than that of HYNIC-3P–RGD2. This is most likely related to the fact that both 99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 are bivalent in binding to integrin αvβ3. As a result, the small difference in their integrin αvβ3 binding affinity was not reflected in tumor uptake of their 99mTc radiotracers. This explanation is also supported by much lower tumor uptake of [99mTc(HYNIC-RGD2)(tricine)(TPPTS)] (99mTc-RGD2), which is only monovalent in binding to integrin αvβ3 despite of the presence of two cyclic RGD moieties in RGD2.31 Bivalency remains the key factor that contributes to the tumor uptake of integrin αvβ3-targeted radiotracers.18
It must be noted that bivalency is largely dependent on integrin αvβ3 receptor density on a specific tissue. In the tumors like xenografted U87MG gliomas, the integrin αvβ3 density is high, and the distance between two neighboring integrin αvβ3 sites is relatively short. Therefore, it is possible for 99mTc-Galacto-RGD2 or 99mTc-3P–RGD2 to be bivalent in binding to integrin αvβ3. In most normal organs, however, the integrin αvβ3 density is relatively low and the distance between two integrin αvβ3 sites is expected to be much longer. In this situation, 99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 may not be bivalent in binding to integrin αvβ3 expressed on normal tissues.
The major difference between 99mTc-Galacto-RGD2 and 99mTc-3P–RGD2 is that two PEG4 groups between the two RGD moieties in 3P–RGD2 are replaced with a pair of SAA, 1,2,3-triazole and PEG2 moieties. This change resulted in a faster radiotracer washout from tumors (Table 1) and normal organs, most likely due to the improved hydrophilicity. Reduction of radiotracer uptake in the intestines, lungs and spleen (Figure 4A) is important for diagnosis of tumors in chest and abdominal regions. The integrin αvβ3 specificity of 99mTc-Galacto-RGD2 was demonstrated in the blocking study (Figure 4B). 99mTc-Galacto-RGD2 was excellent for imaging the xenografted U87MG tumors (Figure 5A). Its high glioma uptake was completely in agreement with the high integrin αvβ3 expression on both U87MG glioma cells (Figure 5B) and tumor neovasculature (Figure 5C). 99mTc-Galacto-RGD2 also had better metabolic stability than 99mTc-3P–RGD2 during hepatobiliary excretion (Figure 6). All these factors make us to believe that 99mTc-Galacto-RGD2 has advantages over 99mTc-3P–RGD2 for imaging tumors and related metastases in the chest and abdominal regions.
In our previous study, it has been clearly demonstrated that many normal organs (particularly intestine, liver, lung, kidneys and spleen) are integrin αvβ3–positive.38 This is consistent with the reduced uptake of 99mTc-Galacto-RGD2 in these organs (Figure 4B) in the presence of co-injected excess RGD2. When part of the radiotracer uptake is blocked in integrin αvβ3–positive organs, it is not surprising to see elevated blood radioactivity accumulation. However, it doesn’t mean that the radioactivity accumulation in these organs is all integrin αvβ3–oriented. For example, the uptake of 99mTc-Galacto-RGD2 in liver and lung is reduced only by 50%, but its intestine uptake was reduced by ~80% of its origin value. This conclusion is further supported by comparing 60-min biodistribution data of 111In(DOTA-3P–RGD2) and 111In(DOTA-3P–RGK2), which shows very low integrin αvβ3 binding affinity (IC50 = 715.8 ± 45.1 nM), but still has significant uptake in the intestine, lung, kidneys and spleen.39
CONCLUSION
In this study, we evaluated 99mTc-Galacto-RGD2 as a new SPECT radiotracer for imaging the xenografted U87MG glioma tumors. We found that 99mTc-Galacto-RGD2 could be prepared in high specific activity, and was able to maintain its solution stability for >24 h post-labeling. Because of its lower uptake in intestines, lungs and spleen, 99mTc-Galacto-RGD2 has advantages over 99mTc-3P–RGD2 for imaging tumors in the chest and abdominal regions. 99mTc-Galacto-RGD2 also has a very high metabolic stability. On the basis of results from this and our previous studies,31–44 we strongly believe that 99mTc-Galacto-RGD2 is an excellent radiotracer for imaging integrin αvβ3-positive tumors and related metastases.
Supplementary Material
Figure 3.
Radio-HPLC chromatograms of 99mTc-Galacto-RGD2 in the kit matrix at 1 and 24 h post-labeling. The radiometric peak at ~13.5 min was from 99mTc-Galacto-RGD2. Its radiochemical purity was always >95% without any chromatographic purification.
ACKNOWLEDGEMETS
This work was supported by Purdue University, the Challenge Research Award from Purdue Cancer Center, the Indiana Clinical and Translational Sciences Institute funded in part by grant Number TR000006 (Clinical and Translational Award) from the National Institutes of Health, National Center for advancing Translational Science, R01 CA115883 (S.L.) from the National Cancer Institute, and KG111333 (Y.Z. and S.L.) from the Susan G. Komen Breast Cancer Foundation.
Footnotes
Supporting Information Available: The HPLC (Figure SI1), ESI-MS (Figure SI2) and NMR spectral data (Tables SI1 and SI2; Figures SI3 – SI8 for DQF-COSY, NOESY, ROESY, TOCSY, HSQC, and HMBC, respectively) are all in word document to illustrate the purity, composition and structure of Galacto-RGD2. These materials are available free of charge at http://pubs.acs.org.
REFERENCES
- 1.Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med. 1995;1:27–31. doi: 10.1038/nm0195-27. [DOI] [PubMed] [Google Scholar]
- 2.Gasparini G, Brooks PC, Biganzoli E, Vermeulen PB, Bonoldi E, Dirix LY, Ranieri G, Miceli R, Cheresh DA. Vascular integrin αvβ3: a new prognostic indicator in breast cancer. Clinical Can. Res. 1998;4:2625–2634. [PubMed] [Google Scholar]
- 3.Sengupta S, Chattopadhyay N, Mitra A, Ray S, Dasgupta S, Chatterjee A. Role of αvβ3 integrin receptors in breast tumor. J. Exp. Clin. Cancer Res. 2001;20:585–590. [PubMed] [Google Scholar]
- 4.Albelda SM, Mette SA, Elder DE, Stewart RM, Damjanovich L, Herlyn M, Buck CA. Integrin distribution in maliganant melanoma: association of the αvβ3 subunit with tumor progression. Cancer Res. 1990;50:6757–6764. [PubMed] [Google Scholar]
- 5.Brooks PC, Clark RA, Cheresh DA. Requirement of vascular integrin αvβ3 for angiogenesis. Science. 1994;264:569–571. doi: 10.1126/science.7512751. [DOI] [PubMed] [Google Scholar]
- 6.Hwang R, Varner JV. The role of integrins in tumor angiogenesis. Hematol. Oncol. Clin. North. Am. 2004;18:991–1006. doi: 10.1016/j.hoc.2004.09.010. [DOI] [PubMed] [Google Scholar]
- 7.Bello L, Francolini M, Marthyn P, Zhang JP, Carroll RS, Nikas DC, Strasser JF, Villani R, Cheresh DA, Black PM. αvβ3 and αvβ5 integrin expression in glioma periphery. Neurosurgery. 2001;49:380–389. doi: 10.1097/00006123-200108000-00022. [DOI] [PubMed] [Google Scholar]
- 8.Jin H, Varner J. Integrins: roles in cancer development and as treatment targets. Br. J. Cancer. 2004;90:561–565. doi: 10.1038/sj.bjc.6601576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zitzmann S, Ehemann V, Schwab M. Arginine-Glycine-Aspartic acid (RGD)-peptide binds to both tumor and tumor endothelial cells in vivo. Cancer Res. 2002;62:5139–5143. [PubMed] [Google Scholar]
- 10.D’Andrea LD, Del Gatto A, Pedone C, Benedetti E. Peptide-based molecules in angiogenesis. Chem. Biol. Drug Des. 2006;67:115–126. doi: 10.1111/j.1747-0285.2006.00356.x. [DOI] [PubMed] [Google Scholar]
- 11.Meyer A, Auremheimer J, Modlinger A, Kessler H. Targeting RGD recognizing integrins: drug development, biomaterial research, tumor imaging and targeting. Curr. Pharm. Design. 2006;12:2723–2747. doi: 10.2174/138161206777947740. [DOI] [PubMed] [Google Scholar]
- 12.Weber WA, Haubner R, Vabuliene E, Kuhnast B, Webster HJ, Schwaiger M. Tumor angiogenesis targeting using imaging agents. Q. J. Nucl. Med. 2001;45:179–182. [PubMed] [Google Scholar]
- 13.Haubner R, Wester HR. Radiolabeled tracers for imaging of tumor angiogenesis and evaluation of anti-angiogenic therapies. Curr. Pharm. Des. 2004;10:1439–1455. doi: 10.2174/1381612043384745. [DOI] [PubMed] [Google Scholar]
- 14.Liu S. Radiolabeled multimeric cyclic RGD peptides as integrin αvβ3-targeted radiotracers for tumor imaging. Mol. Pharm. 2006;3:472–487. doi: 10.1021/mp060049x. [DOI] [PubMed] [Google Scholar]
- 15.Cai W, Chen X. Multimodality imaging of tumor angiogenesis. J. Nucl. Med. 2008;49:113s–128s. doi: 10.2967/jnumed.107.045922. [DOI] [PubMed] [Google Scholar]
- 16.Liu S. Radiolabeled RGD peptides as integrin αvβ3-targeted radiotracers: maximizing binding affinity via bivalency. Bioconjugate Chem. 2009;20:2199–2213. doi: 10.1021/bc900167c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ingrid Dijkgraaf I, Boerman OC. Molecular imaging of angiogenesis with SPECT. Eur. J. Nucl. Med. Mol. Imaging. 2010;37(Suppl 1):S104–S113. doi: 10.1007/s00259-010-1499-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tateishi U, Oka T, Inoue T. Radiolabeled RGD Peptides as Integrin alpha(v)beta3–targeted PET Tracers. Current Medicinal Chemistry. 2012;19:3301–3309. doi: 10.2174/092986712801215937. [DOI] [PubMed] [Google Scholar]
- 19.Haubner R, Wester HJ, Weber WA, Mang C, Ziegler SI, Goodman SL, Senekowitsch-Schmidtke R, Kessler H, Schwaiger M. Noninvasive imaging of αvβ3 integrin expression using 18F-labeled RGD-containing glycopeptide and positron emission tomography. Cancer Res. 2001;61:1781–1785. [PubMed] [Google Scholar]
- 20.Nwe K, Kim Y-S, Milenic DE, Baidoo KE, Brechbiel MW. 111In- and 203Pb-labeled cyclic arginine-glycine-aspartic acid peptide conjugate as an αvβ3 integrin-binding radiotracer. J. Label. Compd. Radiopharm. 2012;55:423–426. doi: 10.1002/jlcr.2965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pohle K, Notni J, Bussemer J, Kessler H, Schwaiger M, Beer AJ. 68Ga-NODAGA-RGD is a suitable substitute for 18F-Galacto-RGD and can be produced with high specific activity in a cGMP/GRP compliant automated process. Nucl. Med. Biol. 2012;39:777–784. doi: 10.1016/j.nucmedbio.2012.02.006. [DOI] [PubMed] [Google Scholar]
- 22.Li Y, Guo J, Tang S, Lang L, Chen X, Perrin DM. One-step and one-pot-two-step radiosynthesis of cyclo-RGD-18F-aryltrifluoroborate conjugates for functional imaging. Am. J. Nucl. Med. Mol. Imaging. 2013;3:44–56. [PMC free article] [PubMed] [Google Scholar]
- 23.Tsiapa I, Loudos G, Varvarigou A, Fragogeorgi E, Psimadas D, Tsotakos T, Xanthopoulos S, Mihailidis D, Bouziotis P, Nikiforidis GC, Kagadis GC. Biological evaluation of an ornithine-modified 99mTc-labeled RGD peptide as an angiogenesis imaging agent. Nucl. Med. Biol. 2013;40:262–272. doi: 10.1016/j.nucmedbio.2012.10.015. [DOI] [PubMed] [Google Scholar]
- 24.Wu Y, Zhang X, Xiong Z, Cheng Z, Fisher DR, Liu S, Gambhir SS, Chen X. MicroPET imaging of glioma integrin αvβ3 expression using 64Cu-labeled tetrameric RGD peptide. J. Nucl. Med. 2005;46:1707–1718. [PubMed] [Google Scholar]
- 25.Wu Z, Li ZB, Chen K, Cai W, He L, Chin FT, Li F, Chen X. MicroPET of tumor integrin αvβ3 expression using 18F-labeled PEGylated tetrameric RGD peptide (18F-FPRGD4) J. Nucl. Med. 2007;48:1536–1544. doi: 10.2967/jnumed.107.040816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Liu Z, Niu G, Shi J, Liu S, Wang F, Chen X. 68Ga-labeled cyclic RGD dimers with Gly3 and PEG4 linkers: promising agents for tumor integrin αvβ3 PET imaging. Eur. J. Nucl. Med. Mol. Imaging. 2009;36:947–957. doi: 10.1007/s00259-008-1045-1. [DOI] [PubMed] [Google Scholar]
- 27.Liu Z, Liu S, Wang F, Chen X. Noninvasive imaging of tumor integrin expression using 18F-labeled RGD dimer peptide with PEG4 linkers. Eur. J. Nucl. Med. Mol. Imaging. 2009;36:1296–1307. doi: 10.1007/s00259-009-1112-2. [DOI] [PubMed] [Google Scholar]
- 28.Dijkgraaf I, Liu S, Kruijtzer JA, Soede AC, Oyen WJ, Liskamp RM, Corstens FH, Boerman OC. Effect of linker variation on the in vitro and in vivo characteristics of an 111In-labeled RGD Peptide. Nucl. Med. Biol. 2007;34:29–35. doi: 10.1016/j.nucmedbio.2006.10.006. [DOI] [PubMed] [Google Scholar]
- 29.Dijkgraaf I, Kruijtzer JA, Liu S, Soede AC, Oyen WJ, Corstens FH, Liskamp RM, Boerman OC. Improved targeting of the αvβ3 integrin by multimerization of RGD peptides. Eur. J. Nucl. Med. Mol. Imaging. 2007;34:267–273. doi: 10.1007/s00259-006-0180-9. [DOI] [PubMed] [Google Scholar]
- 30.Liu S, Edwards DS, Ziegler MC, Harris AR, Hemingway SJ, Barrett JA. 99mTc-Labeling of a hydrazinonictotinamide-conjugated vitronectin receptor antagonist. Bioconjugate Chem. 2001;12:624–629. doi: 10.1021/bc010012p. [DOI] [PubMed] [Google Scholar]
- 31.Wang L, Shi J, Kim YS, Zhai S, Jia B, Zhao H, Liu Z, Wang F, Chen X, Liu S. Improving tumor targeting capability and pharmacokinetics of 99mTc-labeled cyclic RGD dimers with PEG4 linkers. Mol. Pharm. 2009;6:231–245. doi: 10.1021/mp800150r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Shi J, Wang L, Kim YS, Zhai S, Liu Z, Chen X, Liu S. Improving tumor uptake and excretion kinetics of 99mTc-labeled cyclic Arginine-Glycine-Aspartic (RGD) dimers with triglycine linkers. J. Med. Chem. 2008;51:7980–7990. doi: 10.1021/jm801134k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shi J, Wang L, Kim YS, Zhai S, Jia B, Wang F, Liu S. 99mTcO(MAG2-3G3-dimer): A new integrin αvβ3-targeted radiotracer with high tumor uptake and favorable pharmacokinetics. Eur. J. Nucl. Med. Mol. Imaging. 2009;36:1874–1884. doi: 10.1007/s00259-009-1166-1. [DOI] [PubMed] [Google Scholar]
- 34.Shi J, Kim YS, Zhai S, Liu Z, Chen X, Liu S. Improving tumor uptake and pharmacokinetics of 64Cu-labeled cyclic RGD dimers with triglycine and PEG4 linkers. Bioconjugate Chem. 2009;20:750–759. doi: 10.1021/bc800455p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Shi J, Kim YS, Chakraborty S, Jia B, Wang F, Liu S. 2-Mercaptoacetylglycylglycyl (MAG2) as a bifunctional chelator for 99mTc-labeling of cyclic RGD dimers: effects of technetium chelate on tumor uptake and pharmacokinetics. Bioconjugate Chem. 2009;20:1559–1568. doi: 10.1021/bc9001739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chakraborty S, Liu S, Kim YS, Shi J, Zhou Y, Wang F. Evaluation of 111In-labeled cyclic RGD peptides: tetrameric not tetravalent. Bioconjugate Chem. 2011;21:969–978. doi: 10.1021/bc900555q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhou Y, Kim YS, Chakraborty S, Shi J, Gao H, Liu S. 99mTc-Labeled cyclic RGD peptides for noninvasive monitoring of tumor integrin αvβ3 expression. Mol. Imaging. 2011;10:386–397. doi: 10.2310/7290.2011.00006. [DOI] [PubMed] [Google Scholar]
- 38.Shi J, Zhou Y, Chakraborty S, Kim YS, Jia B, Wang F, Liu S. Evaluation of 111In-labeled cyclic RGD peptides: effects of peptide and PEG4 multiplicity on their tumor uptake, excretion kinetics and metabolic stability. Theranostics. 2011;1:322–340. doi: 10.7150/thno/v01p0322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Shi J, Jia B, Kim YS, Chakraborty S, Zhou Y, Wang F, Liu S. Impact of bifunctional chelators on biological properties of 111In-labeled cyclic peptide RGD dimers. Amino Acids. 2011;41:1059–1070. doi: 10.1007/s00726-009-0439-0. [DOI] [PubMed] [Google Scholar]
- 40.Jia B, Liu Z, Zhu Z, Shi J, Jin X, Zhao H, Li F, Liu S, Wang F. Blood clearance kinetics, biodistribution and radiation dosimetry of a kit-formulated integrin αvβ3-selective radiotracer 99mTc-3PRGD2 in non-human primates. Mol. Imaging Biol. 2011;13:730–736. doi: 10.1007/s11307-010-0385-y. [DOI] [PubMed] [Google Scholar]
- 41.Zhou Y, Kim YS, Lu X, Liu S. Evaluation of 99mTc-labeled cyclic RGD dimers: impact of cyclic RGD peptides and 99mTc chelates on biological properties. Bioconjugate Chem. 2012;23:586–595. doi: 10.1021/bc200631g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhou Y, Kim YS, Chakraborty S, Shi J, Gao H, Liu S. 99mTc-labeled cyclic RGD peptides for noninvasive monitoring of tumor integrin αvβ3 expression. Mol. Imaging. 2011;10:386–397. doi: 10.2310/7290.2011.00006. [DOI] [PubMed] [Google Scholar]
- 43.Shao G, Zhou Y, Liu S. Monitoring glioma growth and tumor necrosis with u-SPECT-II/CT for by targeting integrin αvβ3 . Mol. Imaging. 2013;12:39–48. [PubMed] [Google Scholar]
- 44.Zhou Y, Shao G, Liu S. Imaging breast cancer lung metastasis by u-SPECT-II/CT with an integrin αvβ3-targeted radiotracer 99mTc-3P–RGD2 . Theranostics. 2012;2:577–587. doi: 10.7150/thno.4443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ma Q, Ji B, Jia B, Gao S, Ji T, Wang X, Han Z, Zhao G. Differential diagnosis of solitary pulmonary nodules using 99mTc-3P4-RGD2 scintigraphy. Eur. J. Nucl. Med. Mol. Imaging. 2011;38:2145–2152. doi: 10.1007/s00259-011-1901-2. [DOI] [PubMed] [Google Scholar]
- 46.Zhu Z, Miao W, Li Q, Dai H, Ma Q, Wang F, Yang A, Jia B, Jing X, Liu S, Shi J, Liu Z, Zhao Z, Li F. 99mTc-3PRGD2 for integrin receptor imaging of lung cancer: a multicenter study. J. Nucl. Med. 2012;53:716–722. doi: 10.2967/jnumed.111.098988. [DOI] [PubMed] [Google Scholar]
- 47.Haubner R, Kuhnast B, Mang C, Weber WA, Kessler H, Wester HJ, Schwaiger M. [18F]Galacto-RGD: Synthesis, Metabolic Stability, and Radiation Dose Estimates. Bioconjugate Chem. 2004;15:61–69. doi: 10.1021/bc034170n. [DOI] [PubMed] [Google Scholar]
- 48.Walsh JC, Kolb HC. Applications of click chemistry in radiopharmaceutical development. Chimia. 2010;64:29–33. doi: 10.2533/chimia.2010.29. [DOI] [PubMed] [Google Scholar]
- 49.Harris TD, Sworin M, Williams N, Rajopadhye M, Damphousse PR, Glowacka D, Poirier MJ, Yu K. Synthesis of stable hydrazones of a hydrazinonicotinyl-modified peptide for the preparation of 99mTc-labeled radiopharmaceuticals. Bioconjugate Chem. 1998;10:808–814. doi: 10.1021/bc9900237. [DOI] [PubMed] [Google Scholar]
- 50.Shioiri T, Yamada S. Amino acids and peptides. IX. Phosphorus in organic synthesis. IV. Diphenyl phosphorazidate. A new convenient reagent for the peptide synthesis. Chem. Pharm. Bull. (Tokyo) 1974;22:849–854. doi: 10.1248/cpb.22.1398. [DOI] [PubMed] [Google Scholar]
- 51.Shioiri T, Yamada S. Amino acids and peptides. X. Phosphorus in organic synthesis. V. On the mechanism for the peptide synthesis by diphenyl phosphorazidate. Chem. Pharm. Bull. Chem. Pharm. Bull.(Tokyo) 1974;22:855–858. doi: 10.1248/cpb.22.1398. [DOI] [PubMed] [Google Scholar]
- 52.Efthymiou T, Gong W, Desaulniers JP. Chemical architecture and applications of nucleic acid derivatives containing 1,2,3-triazole functionalities synthesized via click chemistry. Molecules. 2012;17:12665–12703. doi: 10.3390/molecules171112665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Doss M, Kolb HC, Zhang JJ, Bélanger MJ, Stubbs JB, Stabin MG, Hostetler ED, Alpaugh RK, Mehren M, Walsh JC, Haka M, Mocharla VP, Yu JQ. Biodistribution and radiation dosimetry of the integrin marker 18F-RGD-K5 determined from whole-body PET/CT in monkeys and humans. J Nucl. Med. 2012;53:787–795. doi: 10.2967/jnumed.111.088955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mamat C, Ramenda T, Wuest FR. Recent applications of click chemistry for the synthesis of radiotracers for molecular imaging. Mini Rev. Org. Chem. 2009;6:21–34. [Google Scholar]
- 55.Best MD. Click chemistry and bioorthogonal reactions: unprecedented selectivity in the labeling of biological molecules. Biochemistry. 2009;48:6571–6584. doi: 10.1021/bi9007726. [DOI] [PubMed] [Google Scholar]
- 56.Wängler C, Maschauer S, Prante O, Schafer M, Schirrmacher R, Bartenstein P, Eisenhut M, Wängler B. Multimerization of cRGD peptides by click chemistry: synthetic strategies, chemical limitations, and influence on biological properties. ChemBioChem. 2010;11:2168–2181. doi: 10.1002/cbic.201000386. [DOI] [PubMed] [Google Scholar]
- 57.Galibert M, Sancey L, Renaudet O, Coll JL, Dumy P, Boturyn D. Application of click-click chemistry to the synthesis of new multivalent RGD conjugates. Org. Biomol. Chem. 2010;8:5133–5138. doi: 10.1039/c0ob00070a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Colombo M, Bianchi A. Click chemistry for the synthesis of RGD-containing integrin ligands. Molecules. 2010;15:178–197. doi: 10.3390/molecules15010178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Trabocchi A, Menchi G, Cini N, Bianchini F, Raspanti S, Bottoncetti A, Pupi A, Calorini L, Guarna A. Click-chemistry-derived triazole ligands of arginine-glycine-aspartate (RGD) integrins with a broad capacity to inhibit adhesion of melanoma cells and both in vitro and in vivo angiogenesis. J. Med. Chem. 2010;53:7119–7128. doi: 10.1021/jm100754z. [DOI] [PubMed] [Google Scholar]
- 60.Kowalczyk W, Mascaraque A, Sanchez-Navarro M, Rojo J, Andreu D. Convergent synthesis of glycodendropeptides by click chemistry approaches. Eur. J. Org. Chem. 2012:4565–4573. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







