A new in vivo model to analyze hepatic metastasis of the human colon cancer cell line HCT116 in NOD/Shi-scid/IL-2Rγnull (NOG) mice by 18F-FDG PET/CT

  • Authors:
    • Kenji Kawai
    • Katsumi Tamura
    • Ikuko Sakata
    • Jiro Ishida
    • Masayoshi Nagata
    • Hideo Tsukada
    • Hiroshi Suemizu
    • Masato Nakamura
    • Yoshiyuki Abe
    • Tsuyoshi Chijiwa
  • View Affiliations

  • Published online on: November 15, 2012     https://doi.org/10.3892/or.2012.2141
  • Pages: 464-468
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Clinically, 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) is useful in the evaluation of various types of human cancers. While PET analysis has been established to evaluate subcutaneous lesions of human cancers in mice, its applications for internal lesions are still being developed. We are currently evaluating new PET approaches for the effective evaluation of in vivo metastatic lesions in the internal organs of small experimental animals. In this study, we analyzed in vivo hepatic metastases of human colonic cancer in immunodeficient mice (NOD/Shi-scid/IL-2Rγnull, NOG) using PET imaging. This new PET approach has been proposed for the evaluation of in vivo metastatic lesions in internal organs. The human colon cancer line HCT116 (1.0x105 and 1.0x106 cells/mouse) was transplanted by intrasplenic injection. 18F-FDG-PET/CT scans were performed 2 weeks after transplantation. After PET/CT scans, histopathological examinations were performed. PET/CT analysis disclosed multiple metastatic foci and increased standardized uptake values (SUV) of FDG in the livers of NOG mice (control, SUVmean 0.450±0.033, SUVmax 0.635±0.017; 1.0x105 cells, 0.853±0.087, 1.254±0.237; 1.0x106 cells, 1.211±0.108, 1.701±0.158). There were significant differences in FDG uptakes between the three groups (ANOVA, P=0.017 in SUVmean; P=0.044 in SUVmax, n=2). We clearly and quantitatively detected images of hepatic metastasis in the livers of NOG mice by 18F-FDG-PET/CT in vivo. PET/CT analysis of internal organ lesions of human cancerous xenografts is a new reliable experimental system to simulate metastases. This model system is useful for analyzing metastatic mechanisms and for developing new novel drugs targeting hepatic metastases of cancer.

Introduction

Chemotherapy has contributed to improvements in the outcome of colorectal cancer (1,2), yet the control of metastatic lesions is crucial for improving outcomes. The metastatic mechanisms of colon cancer are still not known in detail; however, previous studies by us and others have identified the key molecules related to liver metastasis in colon cancer (35), and pathological findings of budding and venous invasion of tumors are known to be important in predicting the outcome of colorectal cancer (6,7). Clinically, 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG-PET/CT) fusion imaging is a useful tool for evaluating the stage, recurrence, outcome, and effectiveness of treatment in human cancers (811). Some studies have reported that PET/CT is useful in colon cancer (1214). However, PET/CT imaging has a limitation in revealing hepatic metastasis due to normal uptake in the liver (15).

Many studies have shown that in vivo studies with subcutaneous xenografts in nude mice and severe combined immunodeficient (SCID) mice are useful for analyzing human cancers (1619), whereas they are limited in evaluating internal organ metastases associated with human cancers. Immunodeficient mice commonly show poor metastatic lesions in vivo. We reported that distant metastatic lesions were easily reproduced by human cancer cell lines in newly developed NOD/Shi-scid/IL-2Rγnull (NOG) mouse models employing systemic injections (2023). It was confirmed that the experimental metastasis model of human cancer using NOG mice was more sensitive and easier to achieve than that using SCID mice due to their multiple immunological dysfunctions not only in cytokine production capability, but also in the functional competence of T, B and NK cells (24,25). We previously developed a reliable new model for assaying hepatic metastasis with superimmunodeficient NOG mice (26,27).

Some in vivo studies have evaluated metastatic lesions of internal organs in mice with conventional modalities such as bio-luminescence imaging (BLI), but difficulties were found in revealing the mechanisms of metastasis (28). Therefore, a new in vivo system to detect internal organ metastatic lesions is required to simulate the clinical situation of metastatic lesions using reliable and quantitative approaches.

In this study, we examined whether 18F-FDG-PET/CT scans could semi-quantitatively reveal abnormal FDG uptakes in hepatic metastasis of human colon cancer cell line HCT116 in NOG mice, thereby overcoming the above described limitations encountered when evaluating metastatic lesions in the liver. Moreover, liver metastasis in NOG mice was studied by histopathological analysis. We showed here that the evaluation of metastatic lesions in NOG mice by PET/CT may be an extremely useful in vivo metastatic model.

Materials and methods

Cell culture

The HCT116 cell line was obtained from the American Type Culture Collection (Manassas, VA, USA) and maintained in McCoy’s 5A medium (Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% heat-inactivated fetal bovine serum, 100 U/ml penicillin and 100 μg/ml streptomycin. Cells were incubated in a humidified (37°C, 5% CO2) incubator and passaged on reaching 80% confluence.

In vivo transplantation of human colon cancer cell line, HCT116

NOG mice (9–12 weeks of age, male) were maintained at the specific pathogen-free facilities of the Central Institute for Experimental Animals (CIEA, Kawasaki, Japan). Experimental liver metastases using NOG mice were generated by intrasplenic injection of HCT116 cells (1.0×105 and 1.0×106/mouse, n=2) and splenectomy. All experiments involving laboratory animals were performed in accordance with the care and use guidelines of the CIEA, according to our previous reports (26,27).

Whole-body imaging of NOG mice with 18F-FDG-PET and CT scans

Positron-emitting fluorine-18 (18F) was produced by 18O(p, n) 18F nuclear reaction using a cyclotron (HM-18; Sumitomo Heavy Industry, Osaka, Japan) at Hamamatsu Photonics PET Center. 18F-FDG-PET and CT scans were performed in NOG mice 14 days after transplantation. The production of 18F-FDG was carried out according to a method described elsewhere (29). The kinetics and distribution patterns of the radiolabeled compound were determined with a small animal PET scanner (ClairvivoPET; Shimadzu Corp., Kyoto, Japan). This scanner consists of depth of interaction (DOI) detector modules with an axial field of view (FOV) of 151 mm, a transaxial FOV of 102 mm, and a transaxial spatial resolution of 1.54 mm in the center (30). Anesthetized mice were placed in a prone position on a fixation plate and then placed in the gantry hole of the PET scanner. After transmission measurement with an external 137Cs point source (22 MBq) for attenuation correction, 18F-FDG at a dose of 6 MBq (0.2 ml) was injected intravenously into each mouse via the tail vein. Data were acquired in list-mode format for 60 min; full 3D sinograms with corrected efficiency, scattering, attenuation, count losses and decay were reconstructed using an iterative 3D dynamic raw-action maximum likelihood algorithm (Drama). Summation images from 40 to 60 min after 18F-FDG injection were reconstructed, and average and maximum values of the standardized uptake (SUVmean, SUVmax) were semi-quantitatively calculated in various organs of NOG mice. After PET scanning, a CT scan was performed with ClairvivoCT (Shimadzu Corp.) in each NOG mouse.

Macroscopic and microscopic examinations

Mice were autopsied after PET/CT scan examinations to evaluate liver metastases. These metastatic lesions were also histologically confirmed. Immunohistochemistry was carried out on 4 μm tissue sections using the Bond Polymer Refine Detection system (Leica Microsystems, Tokyo, Japan) according to the manufacturer’s instructions with minor modifications. In brief, 4 μm sections of formalin-fixed, paraffin-embedded tissues were deparaffinized by Bond Dewax Solution (Leica Microsystems) and an antigen retrieval procedure was carried out using Bond ER solution (Leica Microsystems) for 30 min at 100°C. Endogenous peroxidases were quenched by incubation with hydrogen peroxide for 5 min. Sections were incubated for 30 min at ambient temperate with primary monoclonal antibodies for anti-HLA class 1-A, B, C (Hokudo, Sapporo, Japan) using the biotin-free polymeric horseradish peroxidase (HRP)-linker antibody conjugate system in a Bond-Max automatic slide stainer (Leica Microsystems). Nuclei were counterstained with hematoxylin.

Statistical analysis

Statistical comparisons of data sets were analyzed by a one-way factorial ANOVA. Data are shown as means ± standard error of mean (SEM). These analyses were performed using JMP version 8 software (SAS Institute, Inc., Cary, NC, USA). P-values <0.05 were considered to indicate statistically significant differences.

Results

18F-FDG-PET findings in control NOG mice

18F-FDG- PET/CT showed intense FDG uptakes in the brain (SUVmean 1.519±0.083, SUVmax 2.140±0.127), heart (SUVmean 1.343±0.035, SUVmax 1.868±0.016), kidney (SUVmean 2.163±0.125, SUVmax 3.148±0.150) and bladder (SUVmean 32.986±3.590, SUVmax 49.692±8.152) of control NOG mice (Table I, Fig. 1).

Table I

SUV values in the various organs of control NOG mice by 18F-FDG-PET/CT.

Table I

SUV values in the various organs of control NOG mice by 18F-FDG-PET/CT.

SUVmeanSUVmax
Brain1.519±0.0832.140±0.127
Heart1.343±0.0351.868±0.016
Kidney2.163±0.1253.148±0.150
Bladder32.986±3.59049.692±8.152
Lung0.577±0.0641.384±0.104
Liver0.450±0.0330.635±0.017
Muscle0.151±0.0110.327±0.040
Bone0.214±0.0130.476±0.034
Intestine0.824±0.0332.061±0.111
Testis0.510±0.0221.212±0.335

[i] Standardized uptake values (SUV) were examined by 18F-FDG.

18F-FDG-PET and CT findings of tumor metastasis in NOG mice

18F-FDG-PET/CT showed higher multiple FDG uptakes (SUVmean 0.853±0.087, SUVmax 1.254±0.237) in the livers of NOG mice transplanted with 1.0×105 cells of the HCT116 cell line (Table II, Fig. 2) than those of control NOG mice (SUVmean 0.450±0.033, SUVmax 0.635±0.017). CT scans also showed marked liver swelling in NOG mice transplanted with 1.0×106 cells of HCT116 (Fig. 3). PET/CT showed diffuse higher FDG uptakes (SUVmean 1.211±0.108, SUVmax 1.701±0.158) in the livers of NOG mice with injection of 1.0×106 HCT116 cells than FDG uptakes with 1.0×105 cells. There were significant differences in FDG uptakes between the three groups (ANOVA, P=0.017 in SUVmean, P=0.044 in SUVmax). No other organs with abnormal FDG uptake were noted besides that of the livers of NOG mice transplanted with 1.0×105 and 1.0×106 cells of HCT116.

Table II

SUV values in the livers of NOG mice by 18F-FDG- PET/CT.

Table II

SUV values in the livers of NOG mice by 18F-FDG- PET/CT.

SUVmeanSUVmax
Control 0.450±0.033a 0.635±0.017b
1.0×105 cells of HCT116 0.853±0.087a 1.254±0.237b
1.0×106 cells of HCT116 1.211±0.108a 1.701±0.158b

[i] Standardized uptake values (SUV) were examined by 18F-FDG-PET/CT. ANOVA, aP=0.017, bP=0.044; n=2.

In vivo liver metastasis

Experimental liver metastases, which consisted of solid white masses, were detected in all livers of NOG mice transplanted with 1.0×105 and 1.0×106 cells of the HCT116 cell line. Metastatic hepatomegaly transplanted with 1.0×106 cells was more severe than that with 1.0×105 cells (Fig. 4, H&E). It was also immunohistochemically confirmed that there were obviously more metastatic foci in the livers of NOG mice transplanted with 1.0×106 than in mice transplanted with 1.0×105 HTC116 cells (Fig. 4, HLA).

Discussion

In this study, we clearly and quantitatively demonstrated increased multiple 18F-FDG uptakes in hepatic metastases of human colonic cancer cell line HCT116 in NOG mouse models using a small animal PET/CT system. We previously reported that 18F-FDG-PET/CT clinically is a very useful imaging modality to evaluate human cancers, including rare carcinoma cases (3134). Many reports have shown that animal PET imaging is useful for evaluating human cancer xenografts in vivo. It is usually difficult to reveal metastases as conventional in vivo models are limited in their evaluation of metastases associated with human cancers (28,35). It is easy to produce experimental liver metastases in immunocompromised NOG mice because of their multiple immunological dysfunctions in cytokine production capability as well as functional competence of T, B and NK cells (2427). However, quantitative evaluation of experimental liver metastases is still being developed. We previously reported the use of liver weight as an index of metastatic hepatomegaly or microvessel counts to reflect tumor cell activity (23,36). We present herein that the model system with NOG mice and a small animal PET/CT system has the feasibility of a quantitative metastasis model to simulate clinical situations. Several studies have reported the development of new anticancer therapies and novel drugs for human cancers using animal PET (37,38). This model system presented here with NOG mice and a small animal PET/CT system may be useful for developing new anticancer therapies and novel drugs for hepatic metastases of human cancers.

Although PET imaging with 18F-FDG is routinely used for tumor detection and therapy monitoring, several limitations have been reported with 18F-FDG, such as high uptake in normal tissues of the brain and inflammatory tissues (15,39). In contrast, positron-labeled amino acids and their analogs are considered to be useful because of the low rate of amino acid use in normal control tissues. Natural and unnatural artificial labeled amino acids have been reported (40,41). This hepatic metastasis model, using intrasplenic injection of small numbers of cancer cells into NOG mice, is reliable and quantitative, and more closely mimics in vivo conditions (26). New compounds, which are more sensitive and specific for the detection of human cancers, are expected to replace 18F-FDG (42,43). The hepatic metastasis model system in NOG mice combined with small animal PET/CT analysis will be useful in developing new labeling compounds for PET.

In conclusion, the hepatic metastasis model system in NOG mice combined with PET/CT has the feasibility to simulate clinical situations, and this model system is useful for analyzing mechanisms of metastasis and developing new therapeutic approaches for metastatic lesions of human cancers.

Acknowledgements

We are grateful to Hiroshi Iwata, Takeharu Kaiuchi, Norihiro Harada and Dai Fukumoto for their technical assistance and helpful discussions.

References

1 

O’Connor ES, Greenblatt DY, LoConte NK, et al: Adjuvant chemotherapy for stage II colon cancer with poor prognostic features. J Clin Oncol. 29:3381–3388. 2011.PubMed/NCBI

2 

Karoui M, Roudot-Thoraval F, Mesli F, et al: Primary colectomy in patients with stage IV colon cancer and unresectable distant metastases improves overall survival: results of a multicentric study. Dis Colon Rectum. 54:930–938. 2011. View Article : Google Scholar

3 

Tokunaga T, Oshika Y, Abe Y, et al: Vascular endothelial growth factor (VEGF) mRNA isoform expression pattern is correlated with liver metastasis and poor prognosis in colon cancer. Br J Cancer. 77:998–1002. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Tokunaga T, Nakamura M, Oshika Y, et al: Thrombospondin 2 expression is correlated with inhibition of angiogenesis and metastasis of colon cancer. Br J Cancer. 79:354–359. 1999. View Article : Google Scholar : PubMed/NCBI

5 

Sun L, Hu H, Peng L, et al: P-cadherin promotes liver metastasis and is associated with poor prognosis in colon cancer. Am J Pathol. 179:380–390. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Tanaka M, Hashiguchi Y, Ueno H, Hase K and Mochizuki H: Tumor budding at the invasive margin can predict patients at high risk of recurrence after curative surgery for stage II, T3 colon cancer. Dis Colon Rectum. 46:1054–1059. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Sato T, Ueno H, Mochizuki H, et al: Objective criteria for the grading of venous invasion in colorectal cancer. Am J Surg Pathol. 34:454–462. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Cronin CG, Swords R, Truong MT, et al: Clinical utility of PET/CT in lymphoma. Am J Roentgenol. 194:W91–W103. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Ueda S, Saeki T, Shigekawa T, et al: 18F-fluorodeoxyglucose positron emission tomography optimizes neoadjuvant chemotherapy for primary breast cancer to achieve pathological complete response. Int J Clin Oncol. 17:276–282. 2012. View Article : Google Scholar

10 

Xie L, Saynak M, Veeramachaneni NK, et al: Non-small cell lung cancer: prognostic importance of positive FDG PET findings in the mediastinum for patients with N0–N1 disease at pathologic analysis. Radiology. 261:226–234. 2011.PubMed/NCBI

11 

Abe Y, Tamura K, Sakata I, et al: Clinical implications of 18F-fluorodeoxyglucose positron emission tomography/computed tomography at delayed phase for diagnosis and prognosis of malignant pleural mesothelioma. Oncol Rep. 27:333–338. 2012.

12 

Mori S and Oguchi K: Application of (18)F-fluorodeoxyglucose positron emission tomography to detection of proximal lesions of obstructive colorectal cancer. Jpn J Radiol. 28:584–590. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Ducreux M and Dromain C: Non-invasive imaging tools in colorectal cancer. Rev Prat. 60:1071–1073. 2010.(In French).

14 

Treglia G, Calcagni ML, Rufini V, et al: Clinical significance of incidental focal colorectal (18)F-fluorodeoxyglucose uptake: our experience and a review of the literature. Colorectal Dis. 14:174–180. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Kubota R, Kubota K, Yamada S, Tada M, Ido T and Tamahashi N: Microautoradiographic study for the differentiation of intratumoral macrophages, granulation tissues and cancer cells by the dynamics of fluorine-18-fluorodeoxyglucose uptake. J Nucl Med. 35:104–112. 1994.

16 

Abe Y, Nakamura M, Ohnishi Y, Inaba M, Ueyama Y and Tamaoki N: Multidrug resistance gene (MDR1) expression in human tumor xenografts. Int J Oncol. 5:1285–1292. 1994.PubMed/NCBI

17 

Abe Y, Ohnishi Y, Yoshimura M, et al: P-glycoprotein-mediated acquired multidrug resistance of human lung cancer cells in vivo. Br J Cancer. 74:1929–1934. 1996. View Article : Google Scholar : PubMed/NCBI

18 

Suto R, Abe Y, Nakamura M, et al: P-glycoprotein-mediated acquired multidrug resistance of human osteosarcoma xenografts in vivo. Int J Oncol. 12:287–291. 1998.

19 

Fujimori S, Abe Y, Nishi M, et al: The subunits of glutamate cysteine ligase enhance cisplatin resistance in human non-small cell lung cancer xenografts in vivo. Int J Oncol. 25:413–418. 2004.PubMed/NCBI

20 

Miyakawa Y, Ohnishi Y, Tomisawa M, et al: Establishment of a new model of human multiple myeloma using NOD/SCID/gamma(c)(null) (NOG) mice. Biochem Biophys Res Commun. 313:258–262. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Ikoma N, Yamazaki H, Abe Y, et al: S100A4 expression with reduced E-cadherin expression predicts distant metastasis of human malignant melanoma cell lines in the NOD/SCID/γCnull (NOG) mouse model. Oncol Rep. 14:633–637. 2005.PubMed/NCBI

22 

Hamada K, Monnai M, Kawai K, et al: Liver metastasis models of colon cancer for evaluation of drug efficacy using NOD/Shi-scid IL2Rγnull (NOG) mice. Int J Oncol. 32:153–159. 2008.PubMed/NCBI

23 

Chijiwa T, Abe Y, Ikoma N, et al: Thrombospondin 2 inhibits metastasis of human malignant melanoma through microenvironment-modification in NOD/SCID/γCnull (NOG) mice. Int J Oncol. 34:5–13. 2009.PubMed/NCBI

24 

Ito M, Hiramatsu H, Kobayashi K, et al: NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 100:3175–3182. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Hiramatsu H, Nishikomori R, Heike T, et al: Complete reconstitution of human lymphocytes from cord blood CD34+ cells using the NOD/SCID/gamma(c)(null) mice model. Blood. 102:873–880. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Suemizu H, Monnai M, Ohnishi Y, Ito M, Tamaoki N and Nakamura M: Identification of a key molecular regulator of liver metastasis in human pancreatic carcinoma using a novel quantitative model of metastasis in NOD/SCID/γcnull (NOG) mice. Int J Oncol. 31:741–751. 2007.PubMed/NCBI

27 

Kubo A, Ohmura M, Wakui M, et al: Semi-quantitative analyses of metabolic systems of human colon cancer metastatic xenografts in livers of superimmunodeficient NOG mice. Anal Bioanal Chem. 400:1895–1904. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Deroose CM, De A, Loening AM, et al: Multimodality imaging of tumor xenografts and metastases in mice with combined small-animal PET, small-animal CT, and bioluminescence imaging. J Nucl Med. 48:295–303. 2007.PubMed/NCBI

29 

Oberdorfer F, Hull WE, Traving BC and Maier-Borst W: Synthesis and purification of 2-deoxy-2-[18F]fluoro-D-glucose and 2-deoxy-2-[18F]fluoro-D-mannose: characterization of products by 1H- and 19F-NMR spectroscopy. Int J Rad Appl Instrum A. 37:695–701. 1986.

30 

Mizuta T, Kitamura K, Iwata H, et al: Performance evaluation of a high-sensitivity large-aperture small-animal PET scanner: ClairvivoPET. Ann Nucl Med. 22:447–455. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Ueda S, Tsuda H, Asakawa H, et al: Clinicopathological and prognostic relevance of uptake level using 18F-fluorodeoxyglucose positron emission tomography/computed tomography fusion imaging (18F-FDG PET/CT) in primary breast cancer. Jpn J Clin Oncol. 38:250–258. 2008. View Article : Google Scholar

32 

Abe Y, Tamura K, Sakata I, et al: Usefulness of 18F-FDG positron emission tomography/computed tomography for the diagnosis of pyothorax-associated lymphoma: a report of three cases. Oncol Lett. 1:833–836. 2010.

33 

Abe Y, Tamura K, Sakata I, et al: Unique intense uptake demonstrated by 18F-FDG positron emission tomography/computed tomography in primary pancreatic lymphoma: a case report. Oncol Lett. 1:605–607. 2010.PubMed/NCBI

34 

Ozeki Y, Abe Y, Kita H, et al: A case of primary lung cancer lesion demonstrated by F-18 FDG positron emission tomography/computed tomography (PET/CT) one year after the detection of metastatic brain tumor. Oncol Lett. 2:621–623. 2011.

35 

Takamiya Y, Abe Y, Tanaka Y, et al: Murine P-glycoprotein on stromal vessels mediates multidrug resistance in intracerebral human glioma xenografts. Br J Cancer. 76:445–450. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Hatanaka H, Oshika Y, Abe Y, et al: Vascularization is decreased in pulmonary adenocarcinoma expressing brain-specific angiogenesis inhibitor 1 (BAI1). Int J Mol Med. 5:181–183. 2000.PubMed/NCBI

37 

Pantaleo MA, Nicoletti G, Nanni C, et al: Preclinical evaluation of KIT/PDGFRA and mTOR inhibitors in gastrointestinal stromal tumors using small animal FDG PET. J Exp Clin Cancer Res. 29:1732010. View Article : Google Scholar : PubMed/NCBI

38 

Moroz MA, Kochetkov T, Cai S, et al: Imaging colon cancer response following treatment with AZD1152: a preclinical analysis of [18F]fluoro-2-deoxyglucose and 3′-deoxy-3′-[18F]fluorothymidine imaging. Clin Cancer Res. 17:1099–1110. 2011.PubMed/NCBI

39 

Kubota K, Nakamoto Y, Tamaki N, et al: FDG-PET for the diagnosis of fever of unknown origin: a Japanese multi-center study. Ann Nucl Med. 25:355–364. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Tsukada H, Sato K, Fukumoto D, Nishiyama S, Harada N and Kakiuchi T: Evaluation of D-isomers of O-11C-methyl tyrosine and O-18F-fluoromethyl tyrosine as tumor-imaging agents in tumor-bearing mice: comparison with L- and D-11C-methionine. J Nucl Med. 47:679–688. 2006.PubMed/NCBI

41 

Tsukada H, Sato K, Fukumoto D and Kakiuchi T: Evaluation of D-isomers of O-18F-fluoromethyl, O-18F-fluoroethyl and O-18F-fluoropropyl tyrosine as tumour imaging agents in mice. Eur J Nucl Med Mol Imaging. 33:1017–1024. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Perk LR, Stigter-van Walsum M, Visser GW, et al: Quantitative PET imaging of Met-expressing human cancer xenografts with 89Zr-labelled monoclonal antibody DN30. Eur J Nucl Med Mol Imaging. 35:1857–1867. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Wang H, Liu B, Tian JH, et al: Monitoring early responses to irradiation with dual-tracer micro-PET in dual-tumor bearing mice. World J Gastroenterol. 16:5416–5423. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February 2013
Volume 29 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kawai K, Tamura K, Sakata I, Ishida J, Nagata M, Tsukada H, Suemizu H, Nakamura M, Abe Y, Chijiwa T, Chijiwa T, et al: A new in vivo model to analyze hepatic metastasis of the human colon cancer cell line HCT116 in NOD/Shi-scid/IL-2Rγnull (NOG) mice by 18F-FDG PET/CT. Oncol Rep 29: 464-468, 2013
APA
Kawai, K., Tamura, K., Sakata, I., Ishida, J., Nagata, M., Tsukada, H. ... Chijiwa, T. (2013). A new in vivo model to analyze hepatic metastasis of the human colon cancer cell line HCT116 in NOD/Shi-scid/IL-2Rγnull (NOG) mice by 18F-FDG PET/CT. Oncology Reports, 29, 464-468. https://doi.org/10.3892/or.2012.2141
MLA
Kawai, K., Tamura, K., Sakata, I., Ishida, J., Nagata, M., Tsukada, H., Suemizu, H., Nakamura, M., Abe, Y., Chijiwa, T."A new in vivo model to analyze hepatic metastasis of the human colon cancer cell line HCT116 in NOD/Shi-scid/IL-2Rγnull (NOG) mice by 18F-FDG PET/CT". Oncology Reports 29.2 (2013): 464-468.
Chicago
Kawai, K., Tamura, K., Sakata, I., Ishida, J., Nagata, M., Tsukada, H., Suemizu, H., Nakamura, M., Abe, Y., Chijiwa, T."A new in vivo model to analyze hepatic metastasis of the human colon cancer cell line HCT116 in NOD/Shi-scid/IL-2Rγnull (NOG) mice by 18F-FDG PET/CT". Oncology Reports 29, no. 2 (2013): 464-468. https://doi.org/10.3892/or.2012.2141