Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties

  • Authors:
    • Guopei Luo
    • Jiang Long
    • Xiaobo Cui
    • Zhiwen Xiao
    • Zuqiang Liu
    • Si Shi
    • Liang Liu
    • Chen Liu
    • Jin Xu
    • Min Li
    • Xianjun Yu
  • View Affiliations

  • Published online on: January 18, 2013     https://doi.org/10.3892/ijo.2013.1780
  • Pages: 979-984
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells are thought to be the origin of tumor metastasis. However, evidence of cancer stem cells as the source of lymphatic metastasis in pancreatic cancer is not clear. In this study, we examined the stem cell-like properties of the highly lymphatic metastatic pancreatic cancer cells BxPC-3-LN. Compared with the parental BxPC-3 cells, the BxPC-3-LN cells showed stem cell-like properties, including high lymphatic metastasis potential, self-renewal ability and chemoresistance. In addition, the BxPC-3-LN cells also expressed higher levels of sonic hedgehog and migrating cancer stem cell surface markers (CD133 and CXCR4) compared to the parental BxPC-3 cells. The growth of BxPC-3-LN cells was significantly inhibited by gemcitabine combined with the sonic hedgehog inhibitor cyclopamine. The BxPC-3-LN cells expressed lower levels of let-7, miR-34, miR-107, miR-125, miR-128, miR-130, miR-132 and miR-141 than the parental BxPC-3 cells detected by microRNA PCR array, which were reported to have close relation to stem cell factors. This study provides evidence that cancer stem cells are the major sources of pancreatic cancer lymphatic metastasis, and microRNAs may regulate lymphatic metastasis in pancreatic cancer through modulating cancer stem cells.

Introduction

Pancreatic cancer is a devastating disease with a poor 5-year survival rate of 6% (1). Local recurrences and systematic metastasis are the major reasons for treatment failure, even after curative operation for early stage tumors (24). Lymphatic metastases are one of the common routes for tumor recurrences and metastases in pancreatic cancer and also significantly contribute to its poor prognosis (57). However, the underlying mechanism of lymphatic metastases in pancreatic cancer is not clear (8).

Cancer stem cells (CSCs) are a subpopulation of tumor cells that has the capacity to self-renew (by symmetric and asymmetric division), sustains the heterogeneous lineages of cancer cells and continually maintains tumorigenesis (9). A growing body of evidence has indicated the critical role of CSCs in many solid tumors (1012). In addition to contributing to primary tumor formation, CSCs are also key players in the metastatic processes (12). However, the role of CSCs in pancreatic cancer lymphatic metastasis has not been well elucidated.

We have previously established a pancreatic cancer cell line BxPC-3-LN with aggressive features and highly lymphatic metastatic potential, which can serve as an ideal platform to study the mechanism of lymphatic metastasis in pancreatic cancer (8,13). Given the critical function of CSCs in cancer metastasis, we investigated the stem cell-like properties of BxPC-3-LN cells to confirm the involvement of CSCs in lymphatic metastases of pancreatic cancer. We also examined the expression profile of microRNAs (miRNAs) involved in CSCs and cancer metastasis.

Materials and methods

Cell, tissue culture and animal model

The human pancreatic cancer cell line BxPC-3 was purchased from the American Type Culture Collection (Rockville, MD, USA). Cells were cultured at 37°C in a humidified atmosphere of 95% air and 5% CO2. Eight-week-old immunodeficient male mice (BALB/c nu/nu and NOD/SCID) were obtained from Shanghai SLAC Laboratory Animal Co. Ltd, Shanghai, China. All animal procedures were approved by the Institutional of Animal Care Committee, Fudan University, Shanghai, China.

Lymphatic metastatic potential

The BxPC-3-LN pancreatic cancer cells with highly lymphatic metastatic capability were previously established by consecutive in vivo selection processes (8). Eight-week-old BALB/c mice were orthotopically injected with 1×106/ml cells into the pancreas (n=3). Mice were sacrificed at 6-week end-points to examine lymphatic involvement and metastatic lymph nodes were confirmed by pathological examination. Histological features of primary tumors were examined by hematoxylin and eosin staining.

Flow cytometry

Flow cytometry was used to detect the surface markers of CD133 (Miltenyi Biotec, Bergisch Gladbach, Germany) and CXCR4 (eBioscience, San Diego, CA, USA) by using Beckman Coulter FC500 (Miami, FL, USA). Cells were collected, fixed by resuspensing in 10 ml of 70% ethanol for 30 min and washed in PBS. They were incubated in PBS solution containing CD133 and CXCR4 antibody at 4°C for 40 min. Quantitative values are means ± SEM from 3 independent experiments.

Self-renewal ability

Eight-week-old NOD/SCID mice were injected with 10,000 cells into the right flank (n=3). The formation of primary tumors was examined every week. Mice were sacrificed at 4-week end-points to check tumor formation.

Sphere formation of tumor cells was performed as previously described with a slight modification (14). Cells were maintained in serum-free DMEM/Ham’s F12 medium (Invitrogen, Carlsbad, CA) containing 1% N2 supplement (Invitrogen), 2% B27 supplement (Invitrogen), epidermal growth factor (10 ng/ml; PeproTech), basic fibroblast growth factor (20 ng/ml; PeproTech), heparin (2 μg/ml; Sigma-Aldrich, St. Louis, MO) and plated at a density of 1,000 cells/ml in 6-well ultra-low cluster plates (Corning Incorporated, Corning, NY). Sphere formation was observed under light microscope.

Clone formation ability was examined as previously described (15). In brief, 500 single cells were added to a 6-well culture plate. Culture medium was changed every 3 days. At 2-week end-points, cells were stained with hematoxylin solution. The number of colonies was counted under a light microscope.

Immunofluorescence

Immunofluorescence was performed as previously described (16). In brief, on preincubation with normal blocking serum (diluted 1:20 in PBS) for 30 min, cells were incubated for 1 h with sonic hedgehog (Shh) antibody (Epitomics, Burlingame, CA, USA) at 1:100 dilution. The cells were labeled with an anti-rabbit IgG-PE secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA) at 1:200 dilution. DNA was counterstained with DAPI. Microscope was used to observe the expression of specific markers.

RT-PCR

Shh gene expression was tested by reverse-transcription polymerase chain reaction (RT-PCR). The total RNA of cells was extracted using TRIzol isolation reagent (Invitrogen). The primers used in the PCR reactions were designed using information from the human genomic data base (forward primer 5′-CGGAGCGAGGAAGGGAAAG-3′ and reverse primer 5′-TTGGGGATAAACTGCTTGTAGGC-3′).

Drug cytotoxicity assay

Drug cytotoxicity assay was performed as previously described with a slight modification (8). In brief, the BxPC-3-LN cells were plated at a density of 104 cells per well into 96-multiwell plates and were allowed to grow for 24 h. Cyclopamine (6 μmol/l, Sigma-Aldrich), gemcitabine (100 nM, Eli Lilly) and cyclopamine (6 μmol/l) combined with gemcitabine (100 nM) were added to the cells and then incubated for 48 h at 37°C. After initial incubation, cells were incubated at 37°C for 4 h with 20 μl of MTS (Promega) and absorbance was read at 492 nm. Viability levels are presented as a percentage of the level obtained from the blank control (untreated cells) (mean ± SE, n=6).

MicroRNA PCR array

Isolation of miRNAs from cells was carried out using the mirVana miRNA Isolation Kit (Applied Biosystems). miRNAs expression was quantified by using TaqMan miRNA qRT-PCR assays as previously described (TaqMan Array Human MicroRNA A+B Cards Set v3.0, Applied Biosystems) (17). The PCR reaction was performed on 7900HT Real-Time PCR System (Applied Biosystems) with the following conditions: 16°C 2 min, 42°C 1 min, 50°C 1 sec for 40 cycles. The fold change of miRNAs expression (BxPC-3 vs. BxPC-3-LN) was calculated.

Statistical analysis

Fisher’s exact test and Student’s t-test were applied to compare enumeration data and measure mean t data. Stata 10.0 was used for the tests and p<0.05 was considered statistically significant.

Results

Lymphatic metastasis potential

After 6 weeks of tumor cell implantation orthotopically, the BxPC-3-LN cells generated celiac axis, para-aortic and para-renal lymph node metastases confirmed by pathological examination, with larger primary tumors compared with the parental BxPC-3 cells (Fig. 1A and B). The distribution of metastatic lymph nodes by BxPC-3-LN cells was similar to human disease. No obvious lymphatic metastasis was observed for the parental BxPC-3 cells (Fig. 1A).

BxPC-3-LN cells possess CSC markers

In order to determine whether the lymphatic metastatic BxPC-3-LN cells have CSC-like properties, we examined the expression of the CSC markers CD133+ and CXCR4+ by flow cytometry. As shown in Fig. 2, the BxPC-3-LN cells contained an average of 1.0% of CD133+/CXCR4+ cells, which was 5 times of that in the BxPC-3 cells (0.2%), indicating BxPC-3-LN cells possess CSC-like properties, which may explain the matastatic potential of this cell line.

Self-renewal ability

The self-renewal ability of the BxPC-3-LN and BxPC-3 cells was detected by in vivo tumorigenicity and in vitro sphere and clone formation. The BxPC-3-LN cells generated big tumors with 10,000 cells injected after 4 week of implantation (Fig. 3A right), while no tumor was observed for BxPC-3 cells with the same amount of cells injected (Fig. 3A left). In addition, the BxPC-3-LN cells showed more spheres (Fig. 3B2) and clones formation (Fig. 3C2) than the BxPC-3 cells (Fig. 3B1 and C1).

Shh expression is increased in BxPC-3-LN cells

The expression of Shh in BxPC-3 and the lymphatic metastatic BxPC-3-LN cells was examined by immunofluorescence. The BxPC-3-LN cells showed higher expression of Shh compared with the parental BxPC-3 cells (Fig. 4A). The upregulation of Shh mRNA in the BxPC-3-LN cells was also confirmed by RT-PCR (Fig. 4B). Those data suggest that Shh pathway might be involved in the lymphatic metastasis of pancreatic cancer.

Drug cytotoxicity

MTS assay was used to examine the growth inhibition of cyclopamine, gemcitabine and cyclopamine combined with gemcitabine in BxPC-3 cells. As shown in Fig. 5, cyclopamine combined with gemcitabine (17.3%) caused significant inhibition of cell growth compared with gemcitabine alone (p<0.0001) and cyclopamine alone (p<0.0001) (Fig. 5), indicating that blocking Shh pathway is needed in controling the lymphatic metastasis of pancreatic cancer.

MicroRNA PCR array

Differential expression of miRNAs in the BxPC-3 and BxPC-3-LN cells was quantified by using TaqMan miRNA qRT-PCR assay. Compared with the BxPC-3 cells, several aberrantly expressed miRNAs related to CSCs were found in the BxPC-3-LN cells, including upregulation of miR-572, miR-206, miR-449a, miR-489, miR184 and downregulation of let-7g-3p, let-7i-3p, let-7a-3p, miR-107, miR-128 and miR-141-5p (Fig. 6), suggesting a possible regulatory role of microRNA in lymphatic metastasis of pancreatic cancer.

Discussion

CSC is thought as the major source of tumorigenesis and metastasis (9). CD133 is a stem-like cell surface marker expressed in diverse solid tumors, including pancreatic, brain and colon tumors (12,18,19). In pancreatic cancer, CD133+ expressing cells were associated with tumor initiation, chemoresistance, increased cell migration and invasion (12,20). More importantly, it was demonstrated that a CD133+/CXCR4+ subpopulation was responsible for pancreatic cancer metastasis (12). Previously, we have confirmed the migration, invasive and chemoresistant capability of a lymphatic metastatic pancreatic cancer cell line BxPC-3-LN established by serial in vivo selection (8). In this study, we further demonstrated that BxPC-3-LN cells presented stem cell-like properties, including highly lymphatic metastastic potential, self-renewal ability and chemoresistance. In addition, the BxPC-3-LN cells also expressed higher levels of Shh, which was considered as one of the most important stem cell-related signal pathways (11,12). CD133+/CXCR4+ cells, which were identified as the migrating CSCs in a previous study (12), were found to be enriched in the BxPC-3-LN cells compared with the parental BxPC-3 cells. Cyclopamine (a Shh signal pathway inhibitor) combined with gemcitabine showed greater inhibitory effect on the BxPC-3-LN cells than cyclopamine and gemcitabine alone. Our findings suggest that CD133+/CXCR4+ cells might be the major migrating CSCs and are responsible for the lymphatic metastases of pancreatic cancer.

Previous studies reported that gemcitabine-resistant cells showed cancer stem-like cell phenotype, which underwent epithelial-to-mesenchymal transition (EMT) and showed increased expression of the stem cell markers CD24, CD44 and epithelial specific antigen (ESA) (21,22). These findings indicate that cancer stem-like cells, which are known for their chemoresistant ability, can be enriched during the acquisition of chemoresistance and in therapeutic treatments (23,24). The in vivo serial selection process that was used shares common features to gemcitabine-resistant cells established by exposure to serially escalated doses of gemcitabine, which was also accompanied by an enrichment of CSCs.

miRNAs are a group of short non-coding RNAs that modulate gene expression at the post-transcriptional level, usually leading to translational suppression or gene silencing (25). miRNAs are involved in tumor maintenance, progression, and treatment resistance (2628). There are also evidence showing that miRNAs regulate CSCs by suppressing the expression of ‘stem cell factors’ such as Sox2, Kif4, CD44 and Notch (24,2632). Yu et al(24) found that the CSC-enriched cells expressed low levels of let-7, miR-107, miR-125, miR-128, miR-130, miR-132 and miR-141 compared with the parental cells. Another study showed that restoration of miR-34 strongly suppressed the growth and invasion of p53-mutant pancreatic cancer cells, and sensitized the cells to chemo- and radiotherapy. Moreover, restoration of miR-34 resulted in an 87% decrease in the CD133+/CXCR4+ CSCs by direct regulating its downstream targets Notch1/2 and Bcl-2, suggesting the potential role of miR-34 in CSCs (29). In our study, we also showed that the CSC-enriched BxPC-3-LN cells expressed lower levels of let-7, miR-34, miR-107, miR-125, miR-128, miR-130, miR-132 and miR-141 than the parental BxPC-3 cells, indicating miRNAs regulates the lymphatic metastasis of pancreatic cancer by regulating CSCs. In addition, we found upregulation of miR-184 in BxPC-3-LN cells, which is epigenetically regulated by Methyl-CpG binding protein 1 (MBD1) for modulating stem cell growth and differentiation (33). Our previous studies have demonstrated that MBD1 plays an important role in pancreatic cancer growth and lymphatic metastasis (34,35). These findings suggest that MBD1 regulate pancreatic cancer progression by epigenetic regulation of miR-184, which can affect stem cell properties.

In conclusion, the BxPC-3-LN cells possess stem cell-like properties. The in vivo serial selection processes were similar to gemcitabine-resistant cells established by exposure to serially escalated doses of gemcitabine, which was also accompanied by an enrichment of CSCs. Our results suggest the function of CSCs in pancreatic cancer lymphatic metastasis and miRNAs may regulate pancreatic cancer metastasis through modulating the properties of CSCs.

Acknowledgements

This study was supported in part by the National Science Foundation of China (grant nos. 81172276, 81101807, 81001058, 30901435 and 30972905) (X.Y.), the Shanghai Science and Technology Commission (grant no. 12QH1400600), ‘985 project’ Third Stage Oncology Project (grant no. 985III-YFX0102) (J.L.), the MacDonald Research Fund, and the William and Ella Owens Medical Research Foundation (M.L.). We thank Dr Bo Zhang and Dr Huanyu Xia for their technical assistance.

References

1 

Jemal A, Siegel R, Ward E, Hao Y, Xu J and Thun MJ: Cancer statistics, 2009. CA Cancer J Clin. 59:225–249. 2009. View Article : Google Scholar

2 

Luo G, Long J, Zhang B, et al: Stroma and pancreatic ductal adenocarcinoma: An interaction loop. Biochim Biophys Acta. 1826:170–178. 2012.PubMed/NCBI

3 

Warshaw AL and Fernandez-del Castillo C: Pancreatic carcinoma. N Engl J Med. 326:455–465. 1992. View Article : Google Scholar : PubMed/NCBI

4 

Li X, Ma Q, Xu Q, et al: SDF-1/CXCR4 signaling induces pancreatic cancer cell invasion and epithelial-mesenchymal transition in vitro through non-canonical activation of Hedgehog pathway. Cancer Lett. 322:169–176. 2012. View Article : Google Scholar

5 

Pedrazzoli S, DiCarlo V, Dionigi R, et al: Standard versus extended lymphadenectomy associated with pancreatoduodenectomy in the surgical treatment of adenocarcinoma of the head of the pancreas: a multicenter, prospective, randomized study. Lymphadenectomy Study Group. Ann Surg. 228:508–517. 1998. View Article : Google Scholar

6 

Li Y, Kong D, Ahmad A, Bao B and Sarkar FH: Pancreatic cancer stem cells: Emerging target for designing novel therapy. Cancer Lett. May 20–2012.(Epub ahead of print).

7 

Ni X, Yang J and Li M: Imaging-guided curative surgical resection of pancreatic cancer in a xenograft mouse model. Cancer Lett. 324:179–185. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Long J, Luo G, Liu C, et al: Development of a unique mouse model for pancreatic cancer lymphatic metastasis. Int J Oncol. 41:1662–1668. 2012.PubMed/NCBI

9 

Clarke MF, Dick JE, Dirks PB, et al: Cancer stem cells - perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res. 66:9339–9344. 2006. View Article : Google Scholar

10 

Visvader JE and Lindeman GJ: Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 8:755–768. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Li C, Heidt DG, Dalerba P, et al: Identification of pancreatic cancer stem cells. Cancer Res. 67:1030–1037. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Hermann PC, Huber SL, Herrler T, et al: Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 1:313–323. 2007. View Article : Google Scholar : PubMed/NCBI

13 

Luo G, Yu X, Jin C, et al: LyP-1-conjugated nanoparticles for targeting drug delivery to lymphatic metastatic tumors. Int J Pharm. 385:150–156. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Singh SK, Clarke ID, Terasaki M, et al: Identification of a cancer stem cell in human brain tumors. Cancer Res. 63:5821–5828. 2003.PubMed/NCBI

15 

Shi WD, Meng ZQ, Chen Z, Lin JH, Zhou ZH and Liu LM: Identification of liver metastasis-related genes in a novel human pancreatic carcinoma cell model by microarray analysis. Cancer Lett. 283:84–91. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Giodini A, Kallio MJ, Wall NR, et al: Regulation of microtubule stability and mitotic progression by survivin. Cancer Res. 62:2462–2467. 2002.PubMed/NCBI

17 

Mitchell PS, Parkin RK, Kroh EM, et al: Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 105:10513–10518. 2008. View Article : Google Scholar : PubMed/NCBI

18 

O’Brien CA, Pollett A, Gallinger S and Dick JE: A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 445:106–110. 2007.PubMed/NCBI

19 

Singh SK, Hawkins C, Clarke ID, et al: Identification of human brain tumour initiating cells. Nature. 432:396–401. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Moriyama T, Ohuchida K, Mizumoto K, et al: Enhanced cell migration and invasion of CD133+ pancreatic cancer cells cocultured with pancreatic stromal cells. Cancer. 116:3357–3368. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Shah AN, Summy JM, Zhang J, Park SI, Parikh NU and Gallick GE: Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 14:3629–3637. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Wang Z, Li Y, Kong D, et al: Acquisition of epithelialmesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 69:2400–2407. 2009. View Article : Google Scholar

23 

Hong SP, Wen J, Bang S, Park S and Song SY: CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer. 125:2323–2331. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Yu F, Yao H, Zhu P, et al: let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 131:1109–1123. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Hatfield SD, Shcherbata HR, Fischer KA, Nakahara K, Carthew RW and Ruohola-Baker H: Stem cell division is regulated by the microRNA pathway. Nature. 435:974–978. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Wellner U, Schubert J, Burk UC, et al: The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nat Cell Biol. 11:1487–1495. 2009. View Article : Google Scholar

27 

Liu C, Kelnar K, Liu B, et al: The microRNA miR-34a inhibits prostate cancer stem cells and metastasis by directly repressing CD44. Nat Med. 17:211–215. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Cheng H, Shi S, Cai X, et al: microRNA signature for human pancreatic cancer invasion and metastasis. Exp Ther Med. 4:181–187. 2012.PubMed/NCBI

29 

Ji Q, Hao X, Zhang M, et al: MicroRNA miR-34 inhibits human pancreatic cancer tumor-initiating cells. PLoS One. 4:e68162009. View Article : Google Scholar : PubMed/NCBI

30 

Leal JA and Lleonart ME: MicroRNAs and cancer stem cells: Therapeutic approaches and future perspectives. Cancer Lett. Apr 30–2012.(Epub ahead of print).

31 

Ni X, Long J, Cen P, Chen L, Yang J and Li M: Pancreatic cancer tumour initiating cells: the molecular regulation and therapeutic values. J Cell Mol Med. 16:988–994. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Liu C and Tang DG: MicroRNA regulation of cancer stem cells. Cancer Res. 71:5950–5954. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Liu C, Teng ZQ, Santistevan NJ, et al: Epigenetic regulation of miR-184 by MBD1 governs neural stem cell proliferation and differentiation. Cell Stem Cell. 6:433–444. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Luo G, Jin C, Long J, et al: RNA interference of MBD1 in BxPC-3 human pancreatic cancer cells delivered by PLGA-poloxamer nanoparticles. Cancer Biol Ther. 8:594–598. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Liu C, Chen Y, Yu X, et al: Proteomic analysis of differential proteins in pancreatic carcinomas: Effects of MBD1 knock-down by stable RNA interference. BMC Cancer. 8:1212008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 42 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Luo G, Long J, Cui X, Xiao Z, Liu Z, Shi S, Liu L, Liu C, Xu J, Li M, Li M, et al: Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties. Int J Oncol 42: 979-984, 2013
APA
Luo, G., Long, J., Cui, X., Xiao, Z., Liu, Z., Shi, S. ... Yu, X. (2013). Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties. International Journal of Oncology, 42, 979-984. https://doi.org/10.3892/ijo.2013.1780
MLA
Luo, G., Long, J., Cui, X., Xiao, Z., Liu, Z., Shi, S., Liu, L., Liu, C., Xu, J., Li, M., Yu, X."Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties". International Journal of Oncology 42.3 (2013): 979-984.
Chicago
Luo, G., Long, J., Cui, X., Xiao, Z., Liu, Z., Shi, S., Liu, L., Liu, C., Xu, J., Li, M., Yu, X."Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties". International Journal of Oncology 42, no. 3 (2013): 979-984. https://doi.org/10.3892/ijo.2013.1780