You are on page 1of 6

European Journal of Radiology 79 (2011) 369–374

Contents lists available at ScienceDirect

European Journal of Radiology


journal homepage: www.elsevier.com/locate/ejrad

The synthesis of a d-glucosamine contrast agent, Gd-DTPA-DG, and its


application in cancer molecular imaging with MRI
Wei Zhang a , Yue Chen a,∗ , Da Jing Guo b , Zhan Wen Huang a , Liang Cai a , Ling He c
a
Department of Nuclear Medicine, Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, PR China
b
Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
c
West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Objective: The purpose of this study is to describe the synthesis of Gadolinium-diethylenetriamine pen-
Received 11 July 2010 taacetic acid-deoxyglucosamine (Gd-DTPA-DG) which is a d-glucosamine metabolic MR imaging contrast
Accepted 20 October 2010 agent. We will also discuss its use in a pilot MRI study using a xenograft mouse model of human adeno-
carcinoma.
Keywords: Methods: This novel contrast agent was specifically studied because of its ability to “target” metabolically
Gd-DTPA
active tumor tissues. In this study Gd-DTPA-DG is used to investigate how tumor tissues would react to
Magnetic resonance imaging (MRI)
a dose of 0.2 mmol Gd/kg over a 120 min exposure in a xenograft mouse model. These experiments used
d-Glucosamine
Molecular imaging
athymic mice implanted with human pulmonary adenocarcinoma (A549) as demonstrated by dynamic
Tumor targeting MRI. Alternately, another contrast agent that is not specific for targeting, Gd-DTPA, was used as the
control at a similar dose of gadolinium. Efficacy of the targeted contrast agent was assessed by measuring
relaxation rate in vitro and signal intensity (SI) in vivo. Statistical differences were calculated using one-
way analysis of variance.
Results: The synthesized Gd-DTPA-DG was shown to improve the contrast of tumor tissue in this model.
Gd-DTPA-DG was also shown to have a similar pharmacokinetic rate but generated a higher relaxation
rate in tumor tissues relative to the control contrast Gd-DTPA. In comparison to the pre-contrast imaging,
the SI of tumor tissue in the experimental group was shown to be significantly increased at 15 min after
injection of Gd-DTPA-DG (p < 0.001). The enhanced signal intensity spread from the edge of the tumor to
the center and seemed to strengthen the idea that MRI performance would be useful in different tumor
tissues.
Conclusion: This preliminary study shows that this new chelated contrast agent, Gd-DTPA-DG, can be
specifically targeted to accumulation in tumor tissue as compared to normal tissues. This targeted para-
magnetic contrast agent has potential for specific cancer molecular imaging with MRI.
© 2010 Elsevier Ireland Ltd. All rights reserved.

1. Introduction trol contrast delivery to specific areas of interest using relevant


targeting capabilities [2]. In this emerging field of molecular imag-
The goal of molecular imaging is to noninvasively detect and ing, Gadolinium-diethylenetriamine pentaacetic acid is shown to
characterize nascent pathology based upon a unique presentation be a beneficial MR contrast agent. This agent has a long history of
of biomarkers. These biomarkers should help differentiate abnor- use in MRI. Additionally, several groups have created gadolinium-
mal cells from their normal cell counterparts. MRI is known to containing micelles, microspheres, nanoparticles, and liposomes
have excellent spatial resolution, is reasonably noninvasive, and for the purposes of specified target delivery or for modification of
is a sequentially repeatable process to help track disease changes biodistribution. These gadolinium-containing particles have been
due to its lack of ionizing radiation [1]. However, MRI that is used to increase the site-specific concentration of gadolinium, as
augmented by the use of contrast agents for imaging, can help compared to using aqueous gadolinium or gadolinium-conjugated
further highlight differences between tumor and normal tissues. to targeting molecules [3–5]. We have previously reported a
Contrast agents can be biologically encapsulated in order to con- specifically sensitive nuclear detection agent, technetium-99m
diethylenetriamine pentaacetic acid-d-glucosamine (99m Tc-DTPA-
DG), which showed excellent tumor targeting. These agents
∗ Corresponding author. Tel.: +86 830 3165722; fax: +86 830 3165720. are promising imaging agents for clinical tumor targeting and
E-mail address: chenyue5523@126.com (Y. Chen). imaging. In this study, we explore a mechanism of combining

0720-048X/$ – see front matter © 2010 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.ejrad.2010.10.021
370 W. Zhang et al. / European Journal of Radiology 79 (2011) 369–374

Fig. 1. The synthesis of Gd-DTPA-DG.

gadolinium with DTPA-DG to make a new specific MR contrast 10 mmol) was added to the solution and kept at 80 ◦ C. After
agent. stirring for 2 h, the solution was cooled to room temperature. d-
Glucosamine hydrochloride (2.13 g, 10 mmol) and triethylamine
2. Materials and methods (1.01 g, 10 mmol) was added and the resulting mixture was
stirred for 24 h at room temperature. 80 ml CH2 Cl2 was added
2.1. Materials to the reaction mixture to initiate separation, the supernatant
was then decanted. This pellet crude product was washed with
Chemicals were used without further purification unless stated. dichloromethane (50 ml, 3 times) to remove the remaining DMSO.
DTPA was purchased from Sigma–Aldrich (Shanghai, China), Gd2 O3 This product was then purified by running over a sephadex G-15
was obtained from Rare-Chem. Hi-Tech Co., Ltd. (Huizhou, China). column and the white solid (2.3 g) was obtained. This monosub-
Human adenocarcinoma (A549) cells were purchased from the Key stituted derivative will be used for the next step in the synthesis
Laboratory of Biotherapy of Human Disease of the Ministry of Edu- reaction.
cation, West China Hospital, Sichuan University (Chengdu, Sichuan,
China). RPMI-1640 culture solution, 10% fetal calf serum, paren- 2.2.2.3. The synthesis of Gd-DTPA-DG. The above mentioned mono-
zyme and penicillin-streptomycin were purchased from GIBCO/BRL substituted derivatives (0.554 g, 1 mmol) were dissolved in 8 ml
Corporation (GIBCO Grand Island, NY). Nude mice were purchased water and Gd2 O3 (0.363 g, 1 mmol) was added. After stirring for 6 h
from the Animal Experimental Center of Sichuan University. Gd- at 80 ◦ C, the mixture was filtered to remove excess Gd2 O3 . Acetone
DTPA (Magnevist) was purchased from Schering Plough Corp. (25 ml) was added to the filtrate and the suspension was filtered to
(Schering-Plough Corp., Guangzhou, China). leave a white product. The crude product was washed with acetone
and diethyl ether and after washing yielded 0.612 g of product. A
2.2. Methods diagram of the synthesis of Gd-DTPA-DG is shown in Fig. 1.

2.2.1. Tumor model 2.2.3. In vitro MRI studies


Human pulmonary adenocarcinoma cells were harvested from Consideration of the role of paramagnetically chelated Gd-DTPA
patients with trypsin/EDTA, washed, and suspended in normal was calculated for only T1 , because the T2 values were not statis-
saline at a concentration of 1 × 107 cells/ml. A 0.1 ml dose of sus- tically different at the tested Gd-DTPA particle concentrations, as
pended cells (1 × 106 cells) were injected subcutaneously in the was expected. We therefore only determined T1 relaxivities in vitro.
right dorsal scapular region of mice using a 25-gauge needle. Ani- Gd-DTPA and Gd-DTPA-DG were dissolved in distilled water and
mals were visually inspected every other day for changes in general samples were read using a 1.5-T scanner (Excite-II, Echospeed;
appearance, behavior, and tumor growth. MRI studies were con- GE medical system) accompanied by an 8-channel neurovascu-
ducted when the tumor volume reached 10 mm × 10 mm × 10 mm, lar array coil. Suspended particles containing Gd-DTPA from 0
as measured by caliper. to 0.5 mM were scanned and compared to the same concentra-
tions of unencapsulated Gd-DTPA. For T1 measurements, a fast spin
2.2.2. Preparation of Gd-DTPA-DG echo sequence inversion recovery (FSE IR) sequence was used with
2.2.2.1. The synthesis of DTPA-bis(anhydride). A mixture of DTPA the following imaging parameters: repetition time (TR): 3000 ms;
(7.8 g, 0.02 mol) was added to a 50 ml flask, acetic anhydride (7.6 ml, echo time (TE): 7.8 ms; TI: 50, 75, 100,150, 200, 250, 300, 400,
0.08 mol) and dry pyridine (10 ml, 0.12 mmol) were added and 500, 600, 700, 800, 900 and 1000 ms; band width: 15.6 kHz; FOV:
heated at 65 ◦ C for 24 h. After cooling to room temperature, the 180 mm × 90 mm; slice thickness: 3.0 mm; matrix: 256 × 128; flip
resulting mixture was filtered and the solid portion was washed angle: 90◦ ; and NEX: 1.
with diethyl ether. After drying 15 min under a vacuum, the residue
(6.5 g) was used for the next step in the reaction without extra 2.2.4. In vivo MRI studies
purification. Twenty, Specific Pathogen Free (SPF) female athymic nude mice
(5–6 weeks old, 20–25 g) were randomly assigned to two groups:
2.2.2.2. The synthesis of DTPA-DG. To prepare monosubstituted the experimental group (n = 10) and the control group (n = 10).
derivatives, DTPA-bis (anhydride) (3.57 g, 10 mmol) was dissolved Mice were given vena caudalis injections, where Gd-DTPA-DG was
in 80 ml DMSO by heating at 80 ◦ C. Once dissolved, water (0.18 ml, injected in the experimental group and Gd-DTPA was used in the
W. Zhang et al. / European Journal of Radiology 79 (2011) 369–374 371

control group (0.2 mmol/kg for both groups). The tumor-bearing


mice were anesthetized by intramuscular (im) administration of
a 1:1 mixture of ketamine and midazolam (0.04 ml per mouse),
with 0.8–1.0 l/min O2 sevoflurane as the inhalation anesthetic.
Axial and coronal MR images of the mice were acquired on a
1.5-T scanner (GE Healthcare) using a FSE T1 WI pulse sequence
with a 3 inch surface coil, before injection, and at 15 min, 30 min,
60 min, 90 min, and 120 min after injection of the contrast agent.
Plain and enhanced imaging parameters were as follows: repeti-
tion time (TR): 400 ms; echo time (TE): 14.9 ms; NA: 1; flip angle
(˛): 90◦ ; Matrix: 192 × 128; FOV: 14 mm × 14 mm; slice thickness:
2 mm;and BW: 10 kHz. All data were analyzed using the free soft-
ware package OsiriX (OsiriX Foundation, Switzerland). Regions of
interest (ROIs) were designated in the right dorsal scapular region
in each mouse. Signal intensities (SI) in these ROIs (≥10 mm2 ) were
measured at different time points and intensity measurements
were acquired. The tumor periphery was defined as the outer 2 mm
rim of the measured tumor.

Fig. 2. (A) Representative MR images of Gd-DTPA and Gd-DTPA-DG were visualized


2.2.5. Statistical analysis using test tubes placed in a water bath. These samples show comparisons between
All T1 values and SI values were measured in the experimen- Gd-DTPA and Gd-DTPA-DG with increasing concentrations of Gd and demonstrates
tal and control groups. All data were analyzed using the Statistical SI. (B) Curves are shown demonstrating the Gd-DTPA and Gd DTPA-DG T1 values for
samples in 1.5 ml of distilled water. The graph displays the T1 values for concen-
Package for Social Sciences (SPSS for Windows, Version 14.0, SPSS,
trations of Gd-DTPA and Gd-DTPA-DG which were measured with a fast field-echo
Chicago, IL, USA). Results are expressed as mean ± standard devia- echo-planar inversion recovery MR sequence.
tion (SD). Statistical differences between groups or within groups
of subjects were calculated using repeated-measures or one way
ANOVAs where p < 0.05 is the level of significance.
3.3. In vivo MRI studies

Significant visualization of tumor tissues was observed after the


3. Results injection of both Gd-DTPA-DG and Gd-DTPA. Axial T1 -weighted
images of the tumor enhanced by the Gd-DTPA-DG and Gd-DTPA
3.1. Chemistry are shown in Figs. 3 and 4, respectively. In the control group, the
tumor immediately showed a clear border with uniform visual
The synthesis of Gd-DTPA-DG generates a white solid powder, enhancement. SI of tumor tissues increased over time and reached
high-resolution mass spectrometry (HRMS) and NMR data were a maximum at 15 min. After this point the SI gradually decreased,
generated where m/z (%) for C20 H32 GdN4 O14 [M+H]+ was calcu- and reached baseline after 2 h (p = 0.68). The experimental group
lated at 710.1151. Mass peaks were found at 710.1154. DTPA has showed enhanced intensity in the peripheral region from the begin-
five carboxylate groups (–COOH) and d-glucosamine has an amine ning and after 15 min the signal gradually increased. The visual
group (–NH2 ). These two particles (DTPA and d-glucosamine) were enhancement spread to the tumor center and lasted for 30 min.
then coated over these two kinds of functional groups leading The signal from the central region of the tumor was significantly
Gadolinium to be directly conjugated to the remaining carboxyl increased (p < .001) as compared to the pre-contrast image. The
group. visual enhancement subsequently decreased, but still remained
higher than that of the Gd-DTPA control group at 2 h (p < 0.05).
SI’s of the tumor were measured in both experimental and con-
3.2. In vitro MRI studies trol groups at the baseline and at the subsequent time points, this
graph is shown in Fig. 5.
In this study, the Gd-DTPA-DG caused a change in contrast on
MR images as characterized by their longitudinal relaxation time 4. Discussion
(T1 ) as evaluated by using the 1.5-T scanner. Increased Gd-DTPA-DG
concentration led to a decrease in T1 . Gd-DTPA-DG demonstrated a Gadolinium-diethylenetriamine pentaacetic acid is a positive
shorter T1 than Gd-DTPA at the same concentration, but Gd-DTPA- MRI contrast agent. This agent has a long history of use in MRI exam-
DG and Gd-DTPA had little to no effect on T2 , as was expected at inations, has been FDA-approved for nearly two decades, and is a
this concentration. The highest concentration of Gd-DTPA-DG used well characterized stable chelate. Gd(III) is shown to have a strong
in this study was approximately 0.28 mmol/l, T1 value at this con- paramagnetic effect; among all the elements, it has the greatest
centration was found to be 286.30 ± 0.74 ms, while the T1 value for impact on the relaxation time of the hydrogen proton. Although
the control group (Gd-DTPA) was found to be 335.20 ± 61.57 ms. Gd(III) has some serious and toxic side effects, after chelation with
These T1 values were not significantly different (p = 0.30) from DTPA (diethylenetriamine pentaacetic acid), the complexed ions
each other. At the 0.03–0.07 mmol/l concentration range of the two have only a minor impact on in vivo metabolism of certain enzymes,
particles, T1 values of Gd-DTPA-DG were significantly different as thereby reducing toxicity [6,7]. Gd-DTPA however, is a small-
compared to values for Gd-DTPA (p < 0.001). The smallest concen- molecule ionic contrast agent that has a high osmotic pressure
tration of Gd-DTPA-DG that was used was 0.002 mmol/l, T1 value in vivo and unfavorable pharmacokinetics; it also shows undesir-
for this concentration was measured at 1448.93 ± 2.20 ms, which able background enhancement due to fast extravasation into the
showed a significant difference as compared to distilled water con- extracellular fluid [8]. Most of the small molecule Gd(III) chelate
trol (p < 0.05). The particle-specific T1 values are shown in Fig. 2. is confined to the extracellular regions. As a result, the contrast
372 W. Zhang et al. / European Journal of Radiology 79 (2011) 369–374

Fig. 3. Representative T1 -weighted 2D spin-echo magnetic resonance images of a transverse slice of a mouse bearing A549 tumor; a – precontrast, b – 15 min, c – 30 min, d
– 45 min, e – 90 min and f – 120 min after injection of Gd-DTPA-DG at a dose of 0.2 mmol-Gd/kg.

agent is therefore confined to vascular regions, reducing its abil- molecular probes must be employed as one of the prerequisites for
ity to assist in gathering information about cellular physiology or in vivo molecular imaging [12]. Probes that are amenable to label-
molecular pathology [9,10]. For this reason, new contrast agents ing with radiometals for SPECT or PET can also be chelated using
that target specific organs or body tissues and/or aggregate in spe- Gd(III) for use in MR imaging.
cific regions of tumors are needed [11]. Based on the characteristics of hypermetabolism and rela-
In recent years, contrast agent research, development and appli- tively rapid proliferation, carcinoma cells require a large supply
cations have focused mainly on improving the molecular ligand; in of glucose, much more than that needed by normal cells [13].
general, Gd(III) is still the metal ion of choice. Chemical modifi- This inherent metabolic need could be such that in tumor cell
cation of the ligand skeleton of the chelate can introduce various membranes, there exist a much higher glucose transporter expres-
functional groups that improve the targeting to specific tissues or sion, for example Glut-1 and/or Glut-4. Among all the glucose
organs. Tumor-targeted MR imaging is aimed at implementing the transporters, the Glut-l subtype is expressed in almost all tumor
use of paramagnetic contrast agent aggregation at the site of the cell lines, and consequently is closely identified with tumor
tumor. To achieve targeted MR imaging, appropriately high affinity tissues. Analogs are available that have similar structure to d-

Fig. 4. Representative T1 -weighted 2D spin-echo magnetic resonance images of a transverse slice of a mouse bearing A549 tumor; a – precontrast, b – 15 min, c – 30 min, d
– 45 min, e – 90 min, and f – 120 min after the injection of Gd-DTPA at a dose of 0.2 mmol-Gd/kg.
W. Zhang et al. / European Journal of Radiology 79 (2011) 369–374 373

Noting that contrast agents may easily escape from leaky blood
vessels and infiltrate into the extravascular regions; may lead to
incorporation via glucose transporter into other high metabolically
active cells.

5. Conclusions

We have designed, synthesized, and tested a novel functional


d-glucosamine based contrast agent for MR molecular imaging.
Through MR imaging in vitro and using a human lung carci-
noma xenograft animal model in vivo, the agent yielded excellent
T1 and visual enhancement of the tumor. The imaging of can-
cer structures remained enhanced for 2 h after administration of
Gd-DTPA-DG. Compared to the conventional contrast agent, Gd-
DTPA, Gd-DTPA-DG has a longer retention time in tumor tissue
and better reflects the increased level of tumor cell metabolism
Fig. 5. Representative graph of tumor rim signal intensity (SI) after the injection of as compared to normal tissues. The use of Gd-DTPA-DG may
each contrast agent at a dose of 0.2 mmol-Gd/kg and measured over the time points
allow for early monitoring of primary tumors, discovery of tumor
for the two groups.
metastasis, and offers great promise in the discovery of tumor
recurrence.

glucose and can be transported by Glut-l to Glut-4, at that Conflict of interest statement
time the increased metabolic needs of the tumor cells cause
increased uptake of these analogs. These analogs block the catalytic There is no any conflict of interest.
activity of hexose phosphate isomerase to transform d-glucose-
6-phosphate into 1,6 diphosphate. This blockade in the pathway
Acknowledgements
causes an inability for further metabolism and thus remains in
the cell. Yang et al. [14,15] have generated a glucose deriva-
The authors would like to thank the National Natural Science
tive Ethylenedicysteine-deoxyglucose (EC-DG) and found that it is
Foundation of China for funding this work, 30970858. We are also
involved in cell proliferation and growth activities. This continued
grateful to Dr. Ling He for the synthesis of Gd-DTPA-DG and Dr.
involvement in the cell-cycle pathway would allow for contin-
Da-Jing Guo for his technical support in Magnetic resonance imag-
uous post-treatment follow-ups since glucosamine and glucose
ing.
share the same pathway(s). Whereas glucose may only share 4%
of the hexosamine biosynthetic pathway, glucosamine is possi-
References
bly sharing upwards of 96% of glycolytic/TCA and the hexosamine
pathways. [1] Mulder WJ, der Schaft DW v, Hautvast PA, et al. Early in vivo assessment
Similarly, Chen et al. [16–19] have generated a glucose deriva- of angiostatic therapy efficacy by molecular MRI. FASEB J 2007;21(2):378–
tive through linking the –COOH group of DTPA to the amino group 83.
[2] Doiron AL, Homan KA, Emelianov S, et al. Poly(lactic-co-glycolic) acid as a
of glucosamine. This reaction produced DTPA-DG (diethylenetri- carrier for imaging contrast agents. Pharm Res 2009;26(3):674–82.
amine pentaacetic acid-deoxyglucose). Labeling of DTPA-DG with [3] Lipinski MJ, Amirbekian V, Frias JC, et al. MRI to detect atherosclerosis with
99m Tc could reach 99.2% involvement in the glycolytic/TCA path- gadolinium-containing immunomicelles targeting the macrophage scavenger
receptor. Magn Reson Med 2006;56(3):601–10.
way. After a series of studies, they found that 99m Tc-DTPA-DG
[4] Amirbekian V, Lipinski MJ, Briley-Saebo KC, et al. Detecting and assessing
participates in a variety of tumor cell proliferation mechanisms: macrophages in vivo to evaluate atherosclerosis noninvasively using molecular
it can reflect the activity in the nucleus; it can be used for early MRI. Proc Natl Acad Sci USA 2007;104(3):961–6.
[5] Chen HH, Le VC, Qiu B, et al. MR imaging of biodegradable polymeric micropar-
detection of tumor lesions and also be used to evaluate the efficacy
ticles: a potential method of monitoring local drug delivery. Magn Reson Med
of chemotherapy. Given the results of the Chen study, we decided 2005;53(3):614–20.
to chelate Gd2 O3 with DTPA-DG, thereby synthesizing Gd-DTPA- [6] Tyszka JM, Fraser SE, Jacobs RE. Magnetic resonance microscopy: recent
DG. Catalytic amination is one of the most promising methods to advances and applications. Curr Opin Biotechnol 2005;16(1):93–9.
[7] Zong Y, Guo J, Ke T, et al. Effect of size and charge on pharmacokinetics and
build the C-N key for which a high degree of regional selectivity and in vivo MRI contrast enhancement of biodegradable polydisulfide Gd(III) com-
high enantioselectivity exist. There is very important academic sig- plexes. J Control Release 2006;112(3):350–6.
nificance in the synthesis of some of these natural products and [8] Brasch R, Turetschek K. MRI characterization of tumors and grading angio-
genesis using macromolecular contrast media: status report. Eur J Radiol
other organic compounds which have biological activity. In this 2000;34(3):148–55.
study, we use a simple catalyst to activate functional nitrogen trans- [9] Crich SG, Lanzardo S, Alberti D, et al. Magnetic resonance imaging detection
fer reactions leading to highly regioselective and efficient direct of tumor cells by targeting low-density lipoprotein receptors with Gd-loaded
low-density lipoprotein particles. Neoplasia 2007;9(12):1046–56.
introduction of nitrogen bonding on the deoxy-glucose SP2 C-H [10] Sosnovik DE, Weissleder R. Emerging concepts in molecular MRI. Curr Opin
position. Gd2 O3 is easy to generate as compared to other tradi- Biotechnol 2007;18(1):4–10.
tional synthesis methods, thus making it a relatively inexpensive [11] Endres PJ, MacRenaris KW, Vogt S, et al. Cell-permeable MR contrast
agents with increased intracellular retention. Bioconjug Chem 2008;19(10):
product.
2049–59.
This contrast agent has similar biological characteristics and [12] Kiessling F, Morgenstern B, Zhang C. Contrast agents and applications to assess
molecular structure to 99m Tc-DTPA-DG. In our present study using tumor angiogenesis in vivo by magnetic resonance imaging. Curr Med Chem
2007;14(1):77–91.
a human lung adenocarcinoma xenograft model, we found the
[13] Ak I, Stokkel MP, Pauwels EK. Positron emission tomography with 2-
contrast performance after a bolus injection of Gd-DTPA-DG was [18F]fluoro-2-deoxy-d-glucose in oncology. Part II. The clinical value
greatly enhanced. Gd-DTPA-DG also has a longer retention time in detecting and staging primary tumours. J Cancer Res Clin Oncol
than the more conventional imaging agent Gd-DTPA in tumor tis- 2000;126(10):560–74.
[14] Yang D, Yukihiro M, Yu DF, et al. Assessment of therapeutic tumor response
sue. Additionally, the tumor tissue was more visible from the edge using 99m Tc-ethylenedicysteine-glucosamine. Cancer Biother Radiopharm
to the center after injection with the Gd-DTPA-DG contrast agent. 2004;19(4):443–56.
374 W. Zhang et al. / European Journal of Radiology 79 (2011) 369–374

[15] Yang DJ, Kim CG, Schechter NR, et al. Imaging with 99m Tc ECDG targeted at [17] Chen Y, Xiong Q, Yang X, et al. Noninvasive scintigraphic detection of tumor
the multifunctional glucose transport system: feasibility study with rodents. with 99m Tc-DTPA-deoxyglucose: an experimental study. Cancer Biother Radio-
Radiology 2003;226(2):465–73. pharm 2007;22(3):403–5.
[16] Chen Y, Huang ZW, He L, et al. Synthesis and evaluation of a technetium- [18] Liang J, Chen Y, Huang Z, et al. Early chemotherapy response evaluation in
99m-labeled diethylenetriaminepentaacetate-deoxyglucose complex tumors by 99m Tc-DTPA-DG. Cancer Biother Radiopharm 2008;23(3):363–70.
([99m Tc]-DTPA-DG) as a potential imaging modality for tumors. Appl Radiat [19] Sun YY, Chen Y. Cancer drug development using glucose metabolism radio-
Isot 2006;64(3):342–7. pharmaceuticals. Curr Pharm Des 2009;15(9):983–7.

You might also like