99mTc-labeled estradiol as an estrogen receptor probe: Preparation and preclinical evaluation

99mTc-labeled estradiol as an estrogen receptor probe: Preparation and preclinical evaluation

    99m Tc-labeled estradiol as an estrogen receptor probe: preparation and preclinical evaluation Xiaotian Xia, Hongyan Feng, Chongjiao...

780KB Sizes 0 Downloads 33 Views

    99m

Tc-labeled estradiol as an estrogen receptor probe: preparation and preclinical evaluation Xiaotian Xia, Hongyan Feng, Chongjiao Li, Chunxia Qin, Yiling Song, Yongxue Zhang, Xiaoli Lan PII: DOI: Reference:

S0969-8051(15)00165-1 doi: 10.1016/j.nucmedbio.2015.09.006 NMB 7770

To appear in:

Nuclear Medicine and Biology

Received date: Revised date: Accepted date:

2 May 2015 22 August 2015 8 September 2015

Please cite this article as: Xia Xiaotian, Feng Hongyan, Li Chongjiao, Qin Chunxia, Song Yiling, Zhang Yongxue, Lan Xiaoli, 99m Tc-labeled estradiol as an estrogen receptor probe: preparation and preclinical evaluation, Nuclear Medicine and Biology (2015), doi: 10.1016/j.nucmedbio.2015.09.006

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Title Page Title: 99mTc-labeled estradiol as an estrogen receptor probe: preparation and preclinical evaluation

RI P

T

Running headline: 99mTc-labeled estradiol: an ER probe

Authors: Xiaotian Xia#, Hongyan Feng#, Chongjiao Li, Chunxia Qin, Yiling Song, Yongxue

SC

Zhang*, Xiaoli Lan*

Affiliations: Department of Nuclear Medicine, Union Hospital, Tongji Medical College,

Imaging, Wuhan 430022, China

#

MA NU

Huazhong University of Science and Technology,Hubei Province Key Laboratory of Molecular

ED

Xiaotian Xia and Hongyan Feng contributed equally to the article.

PT

*Corresponding author: Prof. Xiaoli Lan PhD, MD, and Prof. Yongxue Zhang MD

430022, China.

CE

Address: Department of Nuclear Medicine, Wuhan Union Hospital, No. 1277 Jiefang Ave, Wuhan,

AC

Tel.: +86-27-83692633(O), +86-13886193262 (mobile); Fax: +86-27-85726282. E-mail: [email protected] (X. Lan), and [email protected] (Y. Zhang)

Funding: This work was supported by the National Natural Science Foundation of China (No. 30970853, 81071200), the Natural Science Foundation of Hubei Province of China for Distinguished Young Scholars (No. 2010CDA094).

ACCEPTED MANUSCRIPT Abstract

Introduction: Most breast cancers express estrogen receptors (ERs). Noninvasive imaging of ER

99m

Tc-labeled estradiol, with diethylenetriaminepentaacetic acid (DTPA) as a chelating

RI P

probe,

T

expression may be helpful for planning therapy of ER+ tumors. We developed a new ER-binding

ligand, and assessed its targeting ability in vitro and in vivo.

SC

Methods: 3-aminoethyl estradiol was synthesized in two steps from estrone, followed by

99m

Tc

MA NU

labeling. Western blotting and immunofluorescence staining were used to detect ER expression in MCF-7 and MDA-MB-231 breast cancer cells. Saturation binding and specific binding were performed by incubating MCF-7 cells with increasing concentrations of99mTc-DTPA-estradiol.

ED

Cell uptake, efflux, and blocking assays were also performed. To test

99m

Tc-DTPA-estradiol in

vivo, nude mice bearing either MCF-7- (high ER expression) or MDA-MB-231-derived tumors 99m

Tc-DTPA-estradiol, and underwent single-photon

PT

(low ER expression) were injected with

CE

emission-computed tomography (SPECT). Mice injected with excess unlabeled DTPA-estradiol were used as controls. Ex vivo gamma-counting of tissues from normal and tumor-bearing mice

AC

was used to evaluate 99mTc-DTPA-estradiol biodistribution. Results: The radiochemical purity of 99mTc-DTPA-estradiol was 98.3% ± 1.3% with a specific activity of 33.1 ± 1.5 MBq/µmol (n=3). Western blotting and immunofluorescence staining confirmed extensive expression of ERs by the MCF-7 cells, and less extensive expression by MDA-MB-231 cells. There was high binding affinity of

99m

Tc-DTPA-estradiol to MCF-7 cells

with a > 45% specific rate of total cell uptake. SPECT images and the biodistribution study results showed significantly higher uptake by MCF-7 tumors (6.06 ± 0.38 %ID/g) than by MDA-MB-231 tumors (1.57 ± 0.28 %ID/g). Pre-injection of MCF-7 tumor-bearing nude mice with excess

ACCEPTED MANUSCRIPT unlabeled DTPA-estradiol significantly reduced tumor uptake of

99m

Tc-DTPA-estradiol (2.24 ±

0.28 %ID/g), suggesting that 99mTc-DTPA-estradiol specifically targets ERs in tumors.

stability.

99m

T

Tc-DTPA-estradiol can be synthesized with satisfactory labeling efficiency and

RI P

Conclusions: 99m

Tc-DTPA-estradiol specifically targeted ERs in vitro and in vivo with favorable

SC

pharmacokinetics, allowing ER receptor expression assessment with SPECT imaging.

MA NU

Key words: Breast Cancer; Estrogen Receptors; Estradiol; Single-photon Emission-Computed

AC

CE

PT

ED

Tomography

ACCEPTED MANUSCRIPT INTRODUCTION Breast cancer is one of the most common malignancies of women in China and worldwide. It has

RI P

T

a high mortality rate [1]. Early diagnosis is imperative for long-term survival. The estrogen receptor (ER) is a very important biomarker in breast cancer, often guiding treatment planning.

SC

Tumors with higher concentrations of ERs (ER+) tend to respond to anti-estrogen hormone

MA NU

therapy, whereas ER-negative (ER-) tumors require a different treatment approach [2].

Quantitation of ERs is prone to sampling error. In the past decade, emphasis has been placed on the development of nuclear imaging approaches.

18

F and other cyclotron-produced radionuclides

F-labeled estrogen derivative, 16α-18F-17β-estradiol (18F-FES), has been evaluated clinically,

PT

18

ED

have been used to label ER ligands to develop in vivo imaging agents [3-6]. The most successful

with promising results reported for prediction of treatment response to anti-estrogen drugs such as

CE

tamoxifen [7]. Subsequent studies have been directed at developing methods for labeling estrogen 99m

Tc for single-photon emission-computed tomography (SPECT) [8, 9].

AC

imaging agents with

However, these agents have demonstrated suboptimal target tissue selectivity in vivo, possibly as a result of their high lipophilicity or rapid metabolism [2]. The complex chemistry involved in the radiosynthesis of these compounds to obtain high yields and purities has further hampered their development [9-11].

We synthesized and characterized an estradiol analog, labeled with technetium (I)-99m, and chelated it with diethylenetriaminepentaacetic acid (DTPA), a useful chelating agent for lanthanide metal ions that has been used in a multitude of preclinical and clinical

ACCEPTED MANUSCRIPT radiopharmaceuticals. It possesses the requisite chemistry for covalent attachment to targeting vectors such as amino acids[12], peptides [13, 14], proteins [15], and nanoparticles [16, 17], and is

RI P

T

metabolized by the kidneys, while most estrogen receptor probes undergo hepatic metabolism. We hypothesized that DTPA may be suitable to conjugate estradiol, and DTPA-estradiolcould chelate

99m

Tc-DTPA-estradiol, and assessed its targeting ability with ERs in

vitro and in vivo.

MATERIALS AND METHODS

MA NU

radiocompound, named it

SC

radiometallic isotopes for imaging and therapeutic applications. Therefore, we synthesized a

ED

Synthesis and radiolabeling of DTPA-estradiol

PT

Estrone (2.7 g, 10 mmol) was dissolved in acetone (150 mL). NaOH (600 mg, 15 mmol), bromoacetonitrile (Xiyashiji, Chengdu, China) (1.3 mL, 17.3 mmol), and potassium iodide

CE

(Sigma-Aldrich, St. Louis MO, USA) (120 mg, 0.723 mmol) were added. The reaction mixture

AC

was heated under reflux for 4 h. It was then evaporated to dryness and ethyl acetate (150 mL) was added. After washing the mixture with double-distilled water in a separatory funnel, the organic layer was dried over magnesium sulfate and filtered. The ethyl acetate was evaporated under reduced pressure, and the solid product washed with ether on filter paper. The 3-cyanomethyl estrone weighed 2.32 g (yield 75%). We confirmed its structure using proton NMR (1H-NMR) spectroscopy (CDCl3) (Bruker Biospin®, Rheinstetten, Germany).

We added lithium aluminum hydride (Xiyashiji, China) (270 mg, 7.1 mmol in THF) to 3-cyanomethyl estrone (440 mg, 1.42 mmol) in tetrahydrofuran (THF, 25 mL), and the reaction

ACCEPTED MANUSCRIPT mixture was stirred at room temperature for 4 h. After the solvent was evaporated, the solid was dissolved in ethyl acetate and washed with water. The ethyl acetate layer was dried over

RI P

T

magnesium sulfate and filtered. The solvent was again evaporated. The yield of 3-acetonitrile

SC

estradiol was 68%. Its structure was confirmed using 1H-NMR (CDCl3).

Diethylenetriaminepentaacetic dianhydride (DTPAA) (TCI, Shanghai, China) (357.7 mg, 1 mmol)

MA NU

was dissolved in 10 mL of anhydrous dimethylformamide (DMF), and added to 3-acetonitrile estradiol (315 mg, 1 mmol). The mixture was stirred at 60°C for 24 h, then dialyzed with a molecular weight cut-off of 500 Da (MD31®, Spectrum Labs, Los Angeles, CA USA) for 48 h.

ED

After lyophilization, the product weighed 78.2 mg. The resulting white precipitate was the product

PT

DTPA-estradiol. Its structure was confirmed using 1H-NMR (CDCl3). The synthetic scheme of

CE

DTPA-estradiol is shown in Figure 1.

99m

Tc/99Mo generator (Beijing Atom High Tech Co., Ltd., Beijing,

AC

Na99mTcO4 was eluted from a

China). 99mTc-pertechnetate (37 MBq) was added to a vial containing the DTPA-estradiol (0.5 mg, 720 nmol) and tin (II) chloride (Sigma Aldrich) (SnCl2, 100 μg) in 0.5 mL water. The reaction proceeded for 30 min. Then, the product was purified using Sep-Pak C-18 cartridge (Waters, Milford MA, USA). The radiolabeled mixture was passed through the pre-activated column, washed with saline (5 mL), and eluted with methanol (5 mL). Thin-layer chromatography (TLC) silica-gel paper strips (Gelman Sciences, Rossdorf, Germany) was used to achieve radiochemical purity with saline and a mixture of ammonium acetate: methanol (V:V = 4:1) as eluents.

ACCEPTED MANUSCRIPT Stability of 99mTc-DTPA-estradiol Two hundred microliters of labeled DTPA-estradiol was incubated at 37°C in 500 μL of histidine

RI P

T

(1 mM), cysteine (1 mM), phosphate-buffered saline (PBS), and human serum for 1, 3, 6 and 24 h. The radiochemical purity was assessed using a radio-TLC scanner. Each time point and condition

SC

was analyzed in triplicate.

MA NU

Cell lines and cell culture

All cell lines were purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). The human breast carcinoma MCF-7 cell line was cultured in

ED

Minimum Essential Media (MEM®; Gibco, Carlsbad CA, USA) supplemented with 10% fetal

PT

bovine serum (Gibco), 100 U/mL penicillin and 100 μg/mL streptomycin (Beyotime, Shanghai, China). The human breast cancer MDA-MB-231 cell line was maintained in Leibovitz’s L-15

CE

medium containing 10% fetal bovine serum and 1% penicillin/streptomycin with high humidity at

AC

37°C and 5% CO2 for cell culture. Cells were subpassaged in a 1:2 split using 0.25% trypsin/0.02% ethylene diamine tetraacetic acid (EDTA).

Measuring ER expression Immunofluorescence staining was performed. MCF-7 and MDA-MB-231 cells were digested, resuspended, and grown overnight in six-well plates. Cells adhering to the coverslips were fixed in iced acetone for 10 min. For immunofluorescence staining, the fixed cells were rinsed with PBS, blocked with 1% bovine serum albumin (BSA), then incubated with diluted primary antibody (rabbit anti-ER, 1:50) (Epitomics, Abcam, Cambridge, England) overnight at 4°C, followed by

ACCEPTED MANUSCRIPT incubation with diluted secondary antibody (Cy3-labeled goat anti-rabbit IgG, 1:200) (Beyotime, China) for 60 min at 37°C. Finally, the cells were incubated with 4-6-diamidino-2-phenylindole

RI P

T

(DAPI) for 5 min and a quenching-resisting agent was added before observation by confocal

SC

microscopy (Nikon, Osaka, Japan).

Western blotting was performed to detect ER protein expression levels in MCF-7 and

MA NU

MDA-MB-231 tumor extracts. Briefly, equal amounts of total protein were separated by sodium lauryl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride membranes. The membranes were then blocked with 5% skim milk for 1 h at room

ED

temperature. After washing, anti-ER monoclonal antibody, diluted 1:1000 (Epitomics, Abcam),

PT

was incubated with the membranes at 4°C overnight. Actin 1:1000 (Santa Cruz Biotechnology, Santa Cruz, CA, USA) was used as a loading control. The blots were washed with PBS, and then

CE

horseradish peroxidase (HRP)-conjugated secondary antibody 1:3000 (Goodbio, Wuhan, China)

AC

was added to the membranes and incubated for 1 h at room temperature. Immunoreactive bands were detected by enhanced chemiluminescence (ECL; Beyotime, Wuhan, China).

Cell culture Assay Saturation binding experiments were performed as described previously [18, 19]. Approximately 2 × 105 MCF-7 cells were seeded per well and incubated at 37°C for 24 h, followed by incubation with 99mTc-DTPA-estradiol at various concentrations (1–200 nM) at 37°C for 1 h. After washing, the cells were lysed with 1 N NaOH. Radioactivity was measured with a gamma counter (2470, WIZARD®; PerkinElmer, Waltham, MA, USA) and expressed as the percentage of applied

ACCEPTED MANUSCRIPT activity normalized to 2 × 105 cells. The resulting data were used to calculate the efficiency of surface stripping to normalize results obtained from experiments performed at 37°C. Each assay

99m

Tc-DTPA-estradiol were performed on both cell types. After

SC

Cell uptake and efflux studies of

RI P

T

was performed in triplicate.

overnight incubation of 2 × 105 cells in each well of a 24-well plate overnight, the cells were

MA NU

incubated with 99mTc-DTPA-estradiol (4 nmol per well, 0.185 MBq) at 37°C for 60, 120, 180, 240, or 360 min. After removing the supernatant, the cells were washed and then lysed with 1 N NaOH at 37°C for 10 min. The radioactivity of the lysates was measured using an automatic

ED

gamma-counter and the cell uptake rate was expressed as a percentage of the total input

PT

radioactive dose. For the cell efflux study, MCF-7 and MDA-MB-231 cells were incubated with 99m

Tc-DTPA-estradiol at 37°C for 360 min. Following two washings with PBS, the cells were

CE

incubated in culture medium for 60, 120, 180, 240, or 360 min, then collected and counted using

AC

the aforementioned methods.

For the blocking study, MCF-7 cells were incubated with 99mTc-DTPA-estradiol (4 nmol per well, 0.185 MBq) in the presence of no other estradiol, 40 nmol per well unlabeled DTPA-estradiol, or 10-fold estradiol (Xiyashiji, Sichuan, China) (40 nmol per well) at 37°C for 1 h and then treated as before. All tests were performed in triplicate.

Animal models

ACCEPTED MANUSCRIPT BALB/c mice (3–4 weeks) (Experimental Animal Center, Tongji Medical College, Huazhong University of Science and Technology), and BALB/c-nu mice (female, 3–4 weeks) (Beijing HFK

RI P

T

Bioscience Co., Ltd., Beijing, China) were maintained at 22 ± 2°C with an alternating 12-h light/dark cycle. The nude mice were maintained in a barrier system room. All animal experiments

SC

were performed under a protocol approved by the Institutional Animal Care and Use Committee

MA NU

of Tongji Medical College, Huazhong University of Science and Technology.

MCF-7 breast tumor cells (5 × 107 cells) or MDA-MB-231 cells (1 × 107 cells) in 50 μL PBS and 50 μL basement membrane gel (Matrigel®, BD Biosciences, San Jose CA, USA), respectively,

ED

were injected subcutaneously into the right axilla. Mice underwent in vivo SPECT imaging or ex

PT

vivo biodistribution studies when the tumor size reached 0.6-1.2 cm in diameter.

CE

Ex vivo distribution experimentsin healthy and tumor-bearing mice

AC

For tissue distribution experiments, normal female BALB/c mice (n = 5 per group) and MCF-7and MDA-MB-231-tumor-bearing mice(n = 4 per group) were each injected with a 100-µL bolus containing 3.7–7.4 MBq (100–200 µCi, 0.1–0.2 µmol)

99m

Tc-DTPA-estradiol via the tail vein.

Normal animals were sacrificed under deep ketamine-xylazine anesthesia in groups of five at 0.5, 1, 2, 3, and 4 h post-injection. Tumor-bearing animals were sacrificed at 2 and 4 h post-injection. The tumor and organs or tissues (blood, heart, lung, stomach, liver, spleen, kidney, small intestine, large intestine, ovary, bone and muscle) were harvested, rinsed with PBS, wiped with filter paper, and weighed for the gamma-counter measurements. The radioactivity of each sample was measured, decay corrected to the injection time, normalized for injected dose and organ/tissue

ACCEPTED MANUSCRIPT weight, and expressed as percentage of injected dose per gram tissue (%ID/g). Tumor-targeting specificity was evaluated in the blocking group by the administration of a 10-fold excess of 99m

T

Tc-DTPA-estradiol. At 4 h post-injection,

RI P

unlabeled DTPA-estradiol 1 h prior to injection of

the mice were sacrificed and dissected, and the %ID/g was calculated as described above. The

SC

values were corrected for the injected dose and expressed as mean ± standard deviation (SD).

MA NU

SPECT imaging

Nude mice bearing MCF-7 or MDA-MB-231 tumors were injected with

99m

Tc-DTPA-estradiol

(7.4–11.1 MBq, 0.2–0.3 µmol) via the tail vein. For the blocking study, MCF-7 tumor-bearing

ED

mice were administered a 10-fold excess dose of non-labeled DTPA-estradiol 1 h prior to injection 99m

Tc-DTPA-estradiol. SPECT with a pinhole collimator (Symbia T6®; Siemens, Erlangen,

PT

of

Germany) was used for obtaining mouse images. Each anesthetized mouse was placed on the

CE

scanner table in the prone position. SPECT images were acquired at 2 and 4 h post-injection with

AC

an acquisition time of 10 min.

Statistical analysis

Quantitative data are presented as mean ± SD. For statistical classification, a Student’s t-test (2-tailed, unpaired) was performed using commercial software (Prism 5.0®, GraphPad Software, San Diego CA, USA and SPSS 13.0®, IBM, Armonk NY, USA). Differences were statistically significant when P <0.05. Pearson correlation was carried out between different organs for 99m

Tc-DTPA-estradiol.

ACCEPTED MANUSCRIPT RESULTS Precursor identification

RI P

T

The structures of 3-acetonitrile estradiol and DTPA-estradiol were confirmed by 1H-NMR spectroscopy. For 3-acetonitrile estradiol, the 1H-NMR (CDCl3) showed peaks at δ 7.21 (dt, J =

SC

10.8, 5.4 Hz, 1H), 6.71 (td, J = 8.7, 2.6 Hz, 1H), 6.64 (dd, J = 9.1, 2.6 Hz, 1H), 4.20–3.95 (m, 2H), 3.81–3.63 (m, 2H), 3.16–3.02 (m, 1H), 2.90–2.78 (m, 2H), 2.32 (d, J = 13.2 Hz, 1H), 2.24–2.09

MA NU

(m, 2H), 2.00–1.62 (m, 7H), 1.54–1.16 (m, 8H), and 0.79 (s, 3H). Data for DTPA-estradiol (400 MHz, CDCl3-added D2O) showed peaks at: δ 7.21 (dd, 1H), 6.81 (t, 2H), 4.21–4.14 (m, 4H), 3.84–3.44 (m, 8H), 3.06 (s, 2H), 2.79 (m, 4H), 2.41–2.18 (m, 8H), 1.99–1.76 (m, 6H), 1.54–1.36

PT

ED

(m, 6H), and 0.76 (s, 3H).

Radiolabeling identification and stability studies

CE

The radiolabeling yield of 99mTc-DTPA-estradiol was 64.5% ± 2.8%, and the radiochemical purity

AC

was 98.3% ± 1.3% (n=3). The specific activity of

MBq/µmol (n=3). The stability of

99m

Tc-DTPA-estradiol was 33.1 ± 1.5

99m

Tc-DTPA-estradiol was assessed by simultaneous

incubation with 1 mM histidine, 1 mM cysteine, PBS (pH = 7.4), and human serum at 37°C at 1, 3, 6, and 24 h (Figure 2). Little decomposition or dissociation of

99m

Tc-DTPA-estradiol into either

[99mTcO4]− or other subsidiary products was detected under these conditions.

Western blotting and immunofluorescence staining Western blotting analysis of ER expression in MCF-7 and MDA-MB-231 cells is shown in Figure 3A, in which the band at 66 kDa belongs to ER expression. Clear and indistinct bands at 66 kDa

ACCEPTED MANUSCRIPT were seen in MCF-7 and MDA-MB-231 cells, respectively, which suggested that ER was overexpressed in MCF-7 cells but not in MDA-MB-231 cells. In addition, in Figure 3B, a strong

RI P

T

red fluorescence signal distribution was seen from the MCF-7 cells. In contrast, there was almost no red fluorescence signal from the MDA-MB-231 cells, further confirming that MCF-7 cells had

SC

high ER expression while MDA-MB-231 cells had low ER expression. This result suggested that

MA NU

MDA-MB-231 cells could be used as a relative negative control in this study.

In vitro assay

The binding affinity of 99mTc-DTPA-estradiol was also measured by incubating MCF-7 cells with

ED

increasing concentrations (1–200 nM) at 37°C, showing an equilibrium dissociation constant (KD)

PT

of 30.0 ± 4.1 nM (n=3) with a calculated maximal number of binding sites (Bmax) of 2.7 × 105 per cell (Figure 4A). The specific binding of

99m

Tc-DTPA-estradiol varied over the concentrations

AC

CE

tested, ranging from 46.0% ± 4.2% to 90.6% ± 0.2% of total cell-associated uptake.

The blocking study showed that the specific binding of 99mTc-DTPA-estradiol could be blocked in the presence of a 10-fold excess of unlabeled DTPA-estradiol or 10-fold excess of estradiol, in MCF-7 cells (Figure 4B). At 1 h, the tracer uptake rates were only 3.23% ± 0.34% and 3.05% ± 0.23%, which were approximately 51.7% decreased and 54.4% decreased from unblocked conditions (P <0.05), respectively. These data confirm the specificity of binding to ER-positive MCF-7 cells.

99m

Tc-DTPA-estradiol

ACCEPTED MANUSCRIPT The cell uptake study revealed that

99m

Tc-DTPA-estradiol binds strongly to ER-positive MCF-7

cells, and very weakly to MDA-MB-231 cells. For

99m

Tc-DTPA-estradiol, 6.68% ± 0.20% of

RI P

T

probe uptake in MCF-7 cells was measured during the first hour of incubation, followed by peak uptake at 6 h, with 8.07% ± 0.27% probe uptake identified (Figure 4C). The cell efflux study

SC

showed that 99mTc-DTPA-estradiol has good cell retention by MCF-7 cells, with only about 1.99% (decreased from 8.07% to 6.08% of total input radioactivity) of

99m

Tc-DTPA-estradiol efflux

MA NU

observed (Figure 4D). In contrast, significantly lower levels of total radioactivity were observed in both cellular uptake (4.95% ± 0.48%) and retention (4.21% ± 0.24%) during 6-hour incubation

ED

with the MDA-MB-231 cells (Figure 4C and4D).

PT

Biodistribution of 99mTc-DTPA-estradiol in normal and tumor-bearing mice To further evaluate 99mTc-DTPA-estradiol characteristics in vivo, its biodistribution in both tumor

CE

and non-tumor tissues of normal BALB/c mice and tumor-bearing mice was examined (Figure 5,

AC

Tables 1 and 2). The ratios of tumor-to-normal tissues or organs were also calculated (Table 2).

In MCF-7 cell models, tumor uptakes were 4.67 ± 0.39 %ID/g and 6.06 ± 0.38 %ID/g at 2 and 4 h post-injection, respectively (Figure 5A and B). In contrast, uptake within MDA-MB-231 tumors peaked 4 h after injection with 1.57 ± 0.28 %ID/g (P <0.05, Figure 5B). The tumor uptake of labeled DTPA-estradiol in the blocked mice was significantly lower than that in the unblocked mice (2.24 ± 0.28 %ID/g vs 6.06 ± 0.38 %ID/g, P <0.05; Figure 5C). To quantify 99m

Tc-DTPA-estradiol uptake in different tissues and therefore assess its specificity,

tumor-to-normal ratios for tissues and organs were calculated (Table 2). At 4 h post-injection,

ACCEPTED MANUSCRIPT 99m

Tc-DTPA-estradiol showed the highest tumor-to-normal ratios, with 2.93 ± 0.30 for blood and

RI P

T

5.24 ± 1.23 for muscle.

Blood clearance of 99mTc-DTPA-estradiol was 5.14 ± 0.15 %ID/g at 0.5 h and 2.08 ± 0.22 %ID/g

SC

at 4 h. Large amounts of renal and hepatic accumulation (36.10 ± 3.35 %ID/g and 50.47 ± 4.49 %ID/g at 0.5h, 10.07 ± 0.91 %ID/g and 48.52 ± 3.35 %ID/g at 4 h, respectively) were

MA NU

observed, suggesting both renal and hepatic elimination may be involved in the pharmaceutical’s metabolism. Notably, moderate accumulation was also observed in the spleen and ovary.

ED

SPECT imaging

administration of

99m

PT

SPECT images of MCF-7 and MDA-MB-231 tumor-bearing mice were acquired at 2 and 4 h after Tc-DTPA-estradiol, and are shown in Figure 6. MCF-7 tumors were clearly

CE

visible with low background signal as early as 2 h post-injection, compared with little radioactive

AC

accumulation within MDA-MB-231 tumors at any of the tested time points. Under blocking conditions with excess unlabeled DTPA-estradiol, clearly reduced tumor uptake of 99m

Tc-DTPA-estradiol was observed (Figure 6). These results confirm the specificity of

99m

Tc-DTPA-estradiol for ER-positive tumors.

DISCUSSION Advances in molecular and cellular biology are transforming our understanding of basic physiology and pathology; similar advances in molecular imaging technologies now permit dynamic and quantitative studies in vivo with minimal invasiveness [10]. There is a need for an

ACCEPTED MANUSCRIPT accurate, affordable, and most of all, noninvasive method for assessing extramammary estrogen receptor status, which can avoid unnecessary biopsies and permit serial assessments during 99m

T

Tc-based estradiol derivative for ER

RI P

endocrine therapy in breast cancer. We developed a

SC

imaging, and proved its targeting efficiency with in vitro and in vivo imaging.

To meet the need for quantifiable targeting to estrogen receptor sites, we focused on linear

MA NU

polyaminopolycarboxylic acid chelating agents [20]. We intended to enhance the water solubility of the estrogen receptor probe by introducing DTPA groups, expecting to minimize the intrahepatic metabolism. The chelator DTPA was introduced to estradiolvia DTPA anhydride

ED

under mild conditions of pH and temperatures. Previous studies have suggested that 3-acetonitrile

PT

estradiol could be produced via an ER-mediated process, and that 99mTc-labeled estradiol could be a useful agent for imaging ER status [21, 22]. Compared with previous estradiol probes,

CE

DTPA-estradiol is a small molecule with a suitable molecular weight. It does not alter

AC

biodistribution or tumor uptake patterns [21]. In addition, unlike other studies highlighting the difficulty of preparing 99m

99m

Tc-labeled derivatives with high specific activity or yields [9, 11],

Tc-DTPA-estradiol was prepared easily, with a satisfactory yield and labeling efficiency. The

in vitro experiments demonstrated that

99m

Tc-DTPA-estradiol is stable in histidine, cysteine, PBS

and human serum at 37°C for 24 h, with more than 90% remaining intact after 24 h of incubation.

Although we presented specific activity in this study, we could not present the effective specific activity (ESA), which is one of the most important characteristics of a receptor-binding radiopharmaceutical. ESA has often been emphasized for ER radioligands and has been an

ACCEPTED MANUSCRIPT important parameter for routine quality control to validate the synthesis procedures. There are direct and indirect methods for the measurement of ER binding. They are used routinely to

RI P

T

determine relative binding affinity (RBA) and ESA of various cold and radiolabeled ER-binding compounds. Some researchers used scintillation proximity technology to develop a rapid

SC

screening assay for ER ligands with satisfactory results [23]. The assay requires tritiated E2 ([3H]-E2; New England Nuclear, Boston MA, USA). Unfortunately, we were unable to acquire

MA NU

this kit, or the instruments needed to measure tritiated E2, so we are unable to present the ESA.

The binding experiments showed that the binding affinity of

99m

Tc-DTPA-estradiol to ER

ED

receptors was 30.0 ± 4.1 nM, with specific binding resulting in over 45% of total binding.

PT

Classically, estrogen elicits genomic effects on transcription via α and β ERs, which are mainly located in the nucleus. Performing receptor-binding studies on whole cells with steroid analogs

CE

poses challenges related to ligand equilibrium, the lipophilic nature of the ligand, estimation of

AC

free ligand, the processes of receptor activation and degradation, and the presence of multiple estrogen receptor types [24]. As a result, careful consideration is required for data interpretation as one can easily over- or underestimate the KD [10].

99m

Tc-DTPA-estradioldemonstrated a KD

much higher than typically reported values of estradiol itself (0.1–1.0 nM)[25, 26], but was comparable to the affinity of a similar estrogen receptor (11 ± 1.5) nM[10]. Further, 99m

Tc-DTPA-estradiolalso displayed good retention in MCF-7 cells with a maximum of only 1.99%

effluxat 6 h.

SPECT

imaging

and

biodistribution

quantitative

analysis

in

nude

mice

showed

ACCEPTED MANUSCRIPT that99mTc-DTPA-estradiolaccumulated in ER-positive MCF-7 tumors with an uptake range of 4.67 ±0.39 %ID/g – 6.06 ±0.38 %ID/g. The tumor-to-normal ratios peaked at 4 h, with 2.93 ± 0.30 for

contrast,

the

tumor-to-normal

ratios

were

RI P

T

blood and 5.24 ± 1.23 for muscle, allowing easy visualization of the ER-positive tumors. In significantly

in

masses

in

99m

Tc-DTPA-estradiol exhibited

SC

MDA-MB-231-tumor-bearing mice or blocked mice, where

lower

minimal tumor uptake at 1.57 ± 0.28%ID/g and 2.24 ± 0.28%ID/g, respectively.Previous research

MA NU

has shown that estradiol-based estrogen receptor probes can target the ER with good results. GAP-EDL was found to accumulate into ER+ 13762 (rat mammary carcinoma) tumors with a tumor-to-muscle ratio of 7.92 ± 0.56 at 4 h [21]; and 99mTc-estradiol-pyridin-2-yl has been shown

PT

ED

to target MCF-7 tumors with a tumor-to-muscle ratio of 5.67 ± 1.06 at 3 h post-injection [10].

We initially intended to use DTPA to improve the metabolic pathways of the probe to overcome

CE

its high uptake in liver. However, the distribution studies revealed that 99mTc-DTPA-estradiol still

AC

accumulated mainly in the liver, with significant uptake in the spleen, the kidneys and the ovary. Attempts to develop a neutral

99m

Tc-estradiol derivative suitable for imaging of tumors

encountered problems with excessive lipophilicity [27]. Lipophilic compounds are usually characterized by slow elimination from the blood pool, resulting in high accumulation within extensively-perfused organs such as the liver and spleen [8, 9, 11].Additionally, steroid hormones (including estrogen) have been identified as stimulators of the reticuloendothelial system [28, 29], suggesting that phagocytosis within the liver and spleen may be another possible contributor towards 99m

such

a

phenomenon.

We

encountered

similar

problems.

The

uptake

of

Tc-DTPA-estradiol in the liver was 50.47 ± 4.49 %ID/g at 30 min and 48.52 ± 3.35 %ID/g at 4

ACCEPTED MANUSCRIPT h post injection, whereas the kidney, which we expected tobe the critical metabolic organ of the DTPA-chelated probecontained36.10 ± 3.35 %ID/g at 30 min and 10.07 ± 0.91 %ID/g at 4 h. In 99m

T

Tc-DTPA-estradiol for producing

RI P

addition, the ovaries would be expected to accumulate

steroid hormones, especially estradiol [30, 31]. Because this is a preliminary study testing the

SC

imaging performance of this estrogen receptor targeted probe, ovarian accumulation of this probe

MA NU

was not studied, and the estrus cycles of all the female mice were not synchronized.

The major goal for cancer imaging is accurate disease characterization through the application of functional and molecular imaging studies.18F-FES PET cancer imaging has been immensely

ED

valuable to clinical oncologists for staging and visualizing primary and metastatic carcinomas [32].

PT

The supplemental molecular characterization and receptor-expression assessment of the tumor has often assisted in determining endocrine therapy options [7, 33]. As an alternative to PET, we

CE

sought to evaluate the utility of a

99m

Tc-DTPA-chelated estradiol as a SPECT tracer in breast

AC

cancer, which is much less expensive than 18F-FES.

So far, there are no readily available and easily synthesized SPECT agents for assessing reproductive cancers expressing estrogen-binding activity [10]. Despite the challenges and drawbacks, newer

99m

Tc-based estradiol derivatives have been reported, including the one in this

study. However, to date, all of them have failed to provide an alternative to 18F-FES PET, mainly attributable to low tumor uptake compounded with high background levels. To overcome the problems posed by such steroid analogs, the development of neutral nonsteroidal analogs that would specifically bind to each estrogen receptor subtype with high affinity would be a valuable

ACCEPTED MANUSCRIPT step [34, 35].

We developed a novel estradiol-based probe,

99m

Tc-DTPA-estradiol, that specifically binds to the

99m

Tc-DTPA-estradiol showed favorable characteristics in vitro and in vivo,

SC

estrogen receptor.

RI P

T

CONCLUSION

demonstrating potential use in targeted imaging and therapy development in ER-positive tumors.

MA NU

Further studies should be performed to optimize the radiocompound for better pharmacokinetics in vivo, especially decreasing the uptake in the liver.

ED

Acknowledgments

PT

This work was supported by the National Natural Science Foundation of China (No. 30970853, 81071200), the Natural Science Foundation of Hubei Province of China for Distinguished Young

CE

Scholars (No. 2010CDA094). The authors are grateful to Prof. Guoping Yan and Dr. Biao Zhao at

AC

the School of Material Science and Engineering, Wuhan Institute of Technology, for their kind help in preparation of the labeling precursors.

Conflicts of interest The authors declare that they have no conflict of interest.

References [1] Chen W, Zheng R, Zhang S, Zhao P, Zeng H, and Zou X. Report of cancer incidence and mortality in China, 2010.Ann Transl Med 2014;2:61.

ACCEPTED MANUSCRIPT [2] Huang L, Zhu H, Zhang Y, Xu X, Cui W, Yang G, et al. Synthesis and binding affinities of Re(I) and (99m)Tc(I)-containing 16alpha-substituted estradiol complexes: Models for potential breast

RI P

T

cancer imaging agents. Steroids 2010;75:905-11.

[3] LaFrate AL, Carlson KE, and Katzenellenbogen JA. Steroidal bivalent ligands for the estrogen

SC

receptor: design, synthesis, characterization and binding affinities.Bioorg Med Chem 2009;17:3528-35.

MA NU

[4] Seimbille Y, Rousseau J, Benard F, Morin C, Ali H, Avvakumov G, et al. F-18-labeled difluoroestradiols: preparation and preclinical evaluation as estrogen receptor-binding radiopharmaceuticals. Steroids 2002;67:765-75.

ED

[5] Banerjee S, Das T, Chakraborty S, Samuel G, Korde A, Venkatesh M, et al. An

PT

estradiol-conjugate for radiolabelling with 177Lu: an attempt to prepare a radiotherapeutic agent.Bioorg Med Chem 2005;13:4315-22.

CE

[6] Aliaga A, Rousseau JA, Ouellette R, Cadorette J, van Lier JE, Lecomte R, et al. Breast cancer

AC

models to study the expression of estrogen receptors with small animal PET imaging.Nucl Med Biol 2004;31:761-70. [7] Linden HM, Stekhova SA, Link JM, Gralow JR, Livingston RB, Ellis GK, et al. Quantitative fluoroestradiol positron emission tomography imaging predicts response to endocrine treatment in breast cancer.J Clin Oncol 2006;24:2793-9. [8] Bigott HM, Parent E, Luyt LG, Katzenellenbogen JA, and Welch MJ. Design and synthesis of functionalized cyclopentadienyl tricarbonylmetal complexes for technetium-94m PET imaging of estrogen receptors.Bioconjug Chem 2005;16:255-64. [9] Luyt LG, Bigott HM, Welch MJ, and Katzenellenbogen JA. 7alpha- and 17alpha-substituted

ACCEPTED MANUSCRIPT estrogens containing tridentate tricarbonyl rhenium/technetium complexes: synthesis of estrogen receptor imaging agents and evaluation using microPET with technetium-94m.Bioorg Med Chem

RI P

T

2003;11:4977-89.

[10] Nayak TK, Hathaway HJ, Ramesh C, Arterburn JB, Dai D, Sklar LA, et al. Preclinical

SC

development of a neutral, estrogen receptor-targeted, tridentate 99mTc(I)-estradiol-pyridin-2-yl hydrazine derivative for imaging of breast and endometrial cancers.J Nucl Med 2008;49:978-86.

MA NU

[11] Skaddan MB, Wust FR, Jonson S, Syhre R, Welch MJ, Spies H, et al. Radiochemical synthesis and tissue distribution of Tc-99m-labeled 7alpha-substituted estradiol complexes.Nucl Med Biol 2000;27:269-78.

for

Tumor

Imaging

Based

on

Methionine:

PT

SPECT-Radiopharmaceutical

ED

[12] Hazari PP, Shukla G, Goel V, Chuttani K, Kumar N, Sharma R, et al. Synthesis of Specific

Tc-99m-DTPA-bis(methionine).Bioconjug Chem 2010;21:229-39.

CE

[13] Zwanziger D and Beck-Sickinger AG. Radiometal targeted tumor diagnosis and therapy with

AC

peptide hormones.Curr Pharm Design 2008;14:2385-400. [14] Nabuurs RJA, Rutgers KS, Welling MM, Metaxas A, de Backer ME, Rotman M, et al. In Vivo Detection of Amyloid-beta Deposits Using Heavy Chain Antibody Fragments in a Transgenic Mouse Model for Alzheimer's Disease. PloS One 2012;7. [15] Rhodes BA. Direct labeling of proteins with 99mTc. International journal of radiation applications and instrumentation. Part B, Nucl Med Biol 1991;18:667-76. [16] Helbok A, Decristoforo C, Dobrozemsky G, Rangger C, Diederen E, Stark B, et al. Radiolabeling of lipid-based nanoparticles for diagnostics and therapeutic applications: a comparison using different radiometals. J Liposome Res 2010;20:219-27.

ACCEPTED MANUSCRIPT [17] Park JA, Reddy PAN, Kim HK, Kim IS, Kim GC, Chang Y, et al. Gold nanoparticles functionalised by Gd-complex of DTPA-bis(amide) conjugate of glutathione as an MRI contrast

RI P

T

agent.Bioorg Med Chem Lett 2008;18:6135-7.

[18] Ruan W, Sassoon A, An F, Simko JP, and Liu B. Identification of clinically significant tumor

microdissection.Mol Cell Proteomics 2006;5:2364-73.

SC

antigens by selecting phage antibody library on tumor cells in situ using laser capture

MA NU

[19] Roth A, Drummond DC, Conrad F, Hayes ME, Kirpotin DB, Benz CC, et al. Anti-CD166 single chain antibody-mediated intracellular delivery of liposomal drugs to prostate cancer cells.Mol Cancer Ther 2007;6:2737-46.

ED

[20] Varshney R, Sethi SK, Hazari PP, Chuttani K, Soni S, Milton MD, et al. Synthesis of

PT

[DTPA-bis(D-ser)] chelate (DBDSC): an approach for the design of SPECT radiopharmaceuticals based on technetium.Curr Radiopharm 2012;5:348-55.

CE

[21] Takahashi N, Yang DJ, Kohanim S, Oh CS, Yu DF, Azhdarinia A, et al. Targeted functional

AC

imaging of estrogen receptors with 99mTc-GAP-EDL.Eur J Nucl Med Mol Imaging 2007;34:354-62.

[22] Takahashi N, Yang DJ, Kurihara H, Borne A, Kohanim S, Oh CS, et al. Functional imaging of estrogen receptors with radiolabeled-GAP-EDL in rabbit endometriosis model.Acad Radiol 2007;14:1050-7. [23] Beauregard JM, Croteau E, Ahmed N, Ouellette R, van Lier JE, and Benard F. Effective specific activities determined by scintillation proximity counting for production runs of [(18)F]FES and 4F-M[(18)F]FES. Nucl Med Biol 2007;34:325-9. [24] Revankar CM, Cimino DF, Sklar LA, Arterburn JB, and Prossnitz ER. A transmembrane

ACCEPTED MANUSCRIPT intracellular estrogen receptor mediates rapid cell signaling. Science 2005;307:1625-30. [25] Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, et al. Comparison of

RI P

T

the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997;138:863-70.

SC

[26] Hochberg RB and Rosner W. Interaction of 16 alpha-[125I]iodo-estradiol with estrogen receptor and other steroid-binding proteins.Proc Natl Acad Sci U.S.A. 1980;77:328-32.

MA NU

[27] Skaddan MB, Wust FR, and Katzenellenbogen JA. Synthesis and Binding Affinities of Novel Re-Containing 7alpha-Substituted Estradiol Complexes: Models for Breast Cancer Imaging Agents.J Org Chem 1999;64:8108-21.

ED

[28] J.A. P, H.M. C, and N.E.Jr W. Radiopharmaceuticals for genitourinary imaging glomerular

PT

and tubular function anatomy urodynamics and testicular imaging, New York: Macmillan Publishing Company,1990.

CE

[29] Dilworth JR and Parrott SJ. The biomedical chemistry of technetium and rhenium.Chem Soc

AC

Rev 1998;27:43-55.

[30] Drummond AE and Fuller PJ. Ovarian actions of estrogen receptor-beta: an update.Semin Reprod Med 2012;30:32-8. [31] Janik ME, Belkot K, and Przybylo M. Is oestrogen an important player in melanoma progression? Contemp Oncol (Pozn) 2014;18:302-6. [32] Gemignani ML, Patil S, Seshan VE, Sampson M, Humm JL, Lewis JS, et al. Feasibility and predictability of perioperative PET and estrogen receptor ligand in patients with invasive breast cancer.J Nucl Med 2013;54:1697-702. [33] Cintolo JA, Tchou J, and Pryma DA. Diagnostic and prognostic application of positron

ACCEPTED MANUSCRIPT emission tomography in breast imaging: emerging uses and the role of PET in monitoring treatment response.Breast Cancer Res Treat 2013;138:331-46.

RI P

T

[34] Revankar CM, Mitchell HD, Field AS, Burai R, Corona C, Ramesh C, et al. Synthetic estrogen derivatives demonstrate the functionality of intracellular GPR30.Acs Chem Biol

SC

2007;2:536-44.

[35] Bologa CG, Revankar CM, Young SM, Edwards BS, Arterburn JB, Kiselyov AS, et al.

MA NU

Virtual and biomolecular screening converge on a selective agonist for GPR30.Nat Chem Biol

AC

CE

PT

ED

2006;2:207-12.

ACCEPTED MANUSCRIPT Tables

1

2

4

Blood

5.14±0.15

4.23±0.29

3.50±0.19

2.46±0.34

2.08±0.22

Brain

0.21±0.06

0.22±0.08

0.18±0.09

0.18±0.02

0.15±0.04

Heart

1.78±0.09

0.99±0.21

0.67±0.22

0.68±0.05

0.63±0.16

Lung

5.68±0.32

5.03±0.17

3.83±1.12

3.08±0.22

2.53±0.58

Stomach

2.62±0.29

1.69±0.20

1.71±0.55

1.64±0.13

1.35±0.58

Liver

50.47±4.49

62.58±5.40

54.17±5.91

50.19±1.71

48.52±3.35

Spleen

12.04±2.39

10.45±0.67

8.74±1.46

7.25±0.68

6.50±0.65

Kidney

36.10±3.35

21.13±1.07

13.16±2.40

11.51±0.65

10.07±0.91

Small intestine

2.58±0.24

2.26±0.46

2.06±0.55

1.84±0.20

1.69±0.29

Large intestine

4.60±0.27

4.50±0.59

3.67±0.78

3.46±0.08

3.16±0.57

Ovary

2.20±0.35

2.47±0.10

3.06±0.42

3.16±0.21

3.34±0.08

1.93±0.08

1.86±0.38

1.41±0.30

1.38±0.15

1.07±0.16

1.17±0.24

1.15±0.42

1.12±0.15

1.09±0.13

0.98±0.37

Muscle

MA NU

ED

Injected dose (100–200 µCi, 0.1–0.2 µmol per mouse)

AC

*

CE

Bone

SC

3

PT

0.5

RI P

Time (h) Organs

T

Table 1 Biodistribution of 99mTc-DTPA-estradiol in normal BALB/c mice (n = 5, %ID/g ± SD)*

ACCEPTED MANUSCRIPT

Table 2 Biodistribution of 99mTc-DTPA-estradiol and blocking group in nude mice bearing MCF-7 and MDA-MB-231 tumor xenografts (n = 4, %ID/g ± SD)a MDA-MB-231

T

MCF-7

RI P

Organs 4h

Blockingb (4h)

4h

Tumor

4.67±0.39

6.06±0.38

2.24±0.28

1.57±0.28

Blood

3.84±0.36

2.18±0.31

2.54±0.24

2.07±0.16

Brain

0.19±0.06

0.15±0.08

0.14±0.03

0.16±0.09

Heart

0.68±0.13

0.59±0.14

0.47±0.18

0.56±0.14

Lung

3.57±0.26

2.33±0.29

2.07±0.32

2.42±0.26

Stomach

1.81±0.29

1.02±0.22

0.85±0.22

1.09±0.17

46.33±3.25

38.25±5.25

42.38±2.38

6.97±1.05

6.88±1.47

7.19±0.72

59.91±12.22

Spleen

8.13±0.52

Kidney

12.92±1.10

9.57±0.98

10.27±0.39

10.35±1.71

Small intestine

1.76±0.14

1.40±0.14

1.32±0.17

1.45±0.18

Large intestine

3.26±0.45

3.06±0.18

2.13±0.13

2.98±0.40

3.36±0.23

3.56±0.17

2.43±0.25

3.35±0.40

1.20±0.19

1.19±0.25

1.09±0.07

1.22±0.04

1.21±0.17

1.19±0.19

1.09±0.29

0.98±0.13

Muscle

CE

Bone

AC

Ovary

ED

Liver

PT

MA NU

SC

2h

Uptake ratios

Tumor-to-blood

1.22±0.09

2.82±0.39

0.89±0.11

0.76±0.13

Tumor-to-muscle

3.94±0.70

5.24±1.23

2.05±0.12

1.63±0.33

a

Injected dose (100–200 µCi, 0.1–0.2 µmol per mouse) Blocking groupwas performed with 10-fold excess of cold DTPA-estradiol injected 1 h before injection of 99mTc-radiolabeled DTPA-estradiol. b

ACCEPTED MANUSCRIPT Figure Legends

RI P

T

Figure 1 Synthetic scheme of DTPA-estradiol

SC

Figure 2 Stability of 99mTc-DTPA-estradiol incubated at 37°C in 1 mM histidine, 1 mM cysteine,

MA NU

PBS, and human serum, with radiochemical purity assessed by TLC.

Figure 3 MCF-7 and MDA-MB-231 cells were stained with fluorescent anti-ER antibody (red) and DAPI (blue) and analyzed by microscopy (200×). Western blotting and immunofluorescence

ED

staining show positive ER expression by MCF-7 and almost no expression by MDA-MB-231

PT

cells.

99m

CE

Figure 4 In vitro cell saturation binding, specific binding, uptake, efflux, and blocking study of

AC

Tc-DTPA-estradiol. (A) Saturation binding (black diamonds) and specific binding (red squares)

curves were generated by measuring the radioactivity in MCF-7 cells incubated with increasing concentrations of

99m

Tc-DTPA-estradiol as indicated. The inset is the Scatchard plot of the

saturation binding experiment. (B) In blocking studies, cell uptake of

99m

Tc-DTPA-estradiol

conjugates at 1 h was blocked in the presence of 10-fold excess of DTPA-estradiol (1, middle) and 10-fold excess of estradiol (2, right), respectively. (C, D) Cell uptake and cell efflux of 99m

Tc-DTPA-estradiol in MCF-7 and MDA-MB-231 cells were measured at the indicated time

points. All data were determined from three independent experiments and expressed as mean ± SD.

ACCEPTED MANUSCRIPT

Figure 5 Biodistribution of 99mTc-DTPA-estradiol in mouse xenograft tumors. (A) Biodistribution 99m

T

Tc-DTPA-estradiol in nude mice bearing MCF-7 tumors at 2 and 4 h post-injection (n = 4)

RI P

of

was assessed by uptake assay. (B) Tumor uptake of MCF-7 and MDA-MB-231 tumor-bearing

SC

mice at 4 h post-injection. (C) Comparison of tumor uptake in MCF-7 tumor-bearing mice at 4 h

Figure 6 SPECT imaging of

99m

MA NU

post-injection of 99mTc-DTPA-estradiol with/without excess cold DTPA-estradiol.

Tc-DTPA-estradiol in nude mice bearing MCF-7 and

MDA-MB-231 tumors, and blocked tumors with pre-injection of 10-fold excess unlabeled

AC

CE

PT

ED

DTPA-estradiol. Arrows indicate tumors.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 1

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 2

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 3

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 4

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 5

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Figure 6