Aldehyde dehydrogenasehigh gastric cancer stem cells are resistant to chemotherapy

  • Authors:
    • Shimpei Nishikawa
    • Masamitsu Konno
    • Atsushi Hamabe
    • Shinichiro Hasegawa
    • Yoshihiro Kano
    • Katsuya Ohta
    • Takahito Fukusumi
    • Daisuke Sakai
    • Toshihiro Kudo
    • Naotsugu Haraguchi
    • Taroh Satoh
    • Shuji Takiguchi
    • Masaki Mori
    • Yuichiro Doki
    • Hideshi Ishii
  • View Affiliations

  • Published online on: February 22, 2013     https://doi.org/10.3892/ijo.2013.1837
  • Pages: 1437-1442
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells (CSCs) are known to influence chemoresistance, survival, relapse and metastasis. Aldehyde dehydrogenase (ALDH) functions as an epithelial CSC marker. In the present study, we investigated the involvement of ALDH in gastric CSC maintenance, chemoresistance and survival. Following screening for eight candidate markers (CD13, CD26, CD44, CD90, CD117, CD133, EpCAM and ALDH), five gastric cancer cell lines were found to contain small subpopulations of high ALDH activity (ALDHhigh cells). We also examined the involvement of ALDHhigh cell populations in human primary tumor samples. Immunodeficient NOD/SCID mice were inoculated with tumor tissues obtained from surgical specimens. ALDHhigh cells were found to persist in the xenotransplanted primary tumor samples. in the immunodeficient mice, ALDHhigh cells exhibited a greater sphere‑forming ability in vitro and tumorigenic potential in vivo, compared with subpopulations of low ALDH activity (ALDHlow cells). Cell cultures treated with 5-fluoro­uracil and cisplatin exhibited higher numbers of ALDHhigh cells. Notch1 and Sonic hedgehog (Shh) expression was also found to increase in ALDHhigh cells compared with ALDHlow cells. Therefore, it can be concluded that ALDH generates chemoresistance in gastric cancer cells through Notch1 and Shh signaling, suggesting novel treatment targets.

Introduction

The discovery of cancer stem cells (CSCs) in hematopoietic malignancies (1) has revealed that tumor tissues comprise a bulk of proliferating or differentiated tumor cells derived from small populations of self-renewing cells (2). Since their identification in leukemia, CSCs have been detected in solid tumors of the head and neck (3), gastrointestinal system (4), colon (5,6), breast (7) and brain (8,9). CSCs are tumorigenic, which is evident from xenotransplantation in immunodeficient mice, and are resistant to chemoradiation, whereas daughter cells are chemoradiation-sensitive (10,11). Recent studies have demonstrated that CSCs survive chemo- and radiation therapy in hypoxic regions of tumors (10,11). Studies of cell- autonomous mechanisms have revealed the involvement of anaerobic glycolysis in CSC maintenance and chemoradiation resistance (10,11). For example, CD13/aminopeptidase N, a liver CSC marker, regulates reactive oxygen species (ROS) through recycling reduced glutathione (GSH), thus contributing to intracellular ROS decrease following chemoradiation exposure (12). Similarly, intracellular ROS are suppressed after chemoradiation therapy through the activity of the hyaluronic acid receptor, CD44, an adhesion molecule expressed in cancer stem-like cells that directly interacts with pyruvate kinase M2, which is putatively involved in anaerobic glycolysis in CSCs (13). Furthermore, the CD44 variant (CD44v) has been shown to interact with xCT, a glutamate-cystine transporter, and to control intracellular GSH levels (14). CD44 abrogation has been shown to cause a loss of xCT from the cell surface, to suppress tumor growth in a transgenic gastric cancer (GC) mouse model and stimulate the p38 (mitogen- activated protein kinase) pathway (a downstream target of ROS) and the expression of the cell cycle inhibitor, p21(CIP1/WAF1), suggesting that CD44 plays a role in GSH synthesis and protection against ROS in gastrointestinal cancers (14). Taken together, these data indicate that cancer metabolism is critical for the initiation and progression of gastrointestinal CSCs.

In the present study, we investigated cell surface markers in gastric CSCs and after screening eight candidate markers (CD13, CD26, CD44, CD90, CD117, CD133, EpCAM and ALDH), we confirmed the involvement of aldehyde dehydrogenase (ALDH) in sphere formation, tumorigenicity and chemoresistance. Throughout the study of the ALDH pathway, a cancer metabolism regulator, we encountered stemness genes, suggesting novel molecular therapeutic targets.

Materials and methods

Cell lines and cell culture

The human GC cell lines, AGS, NUGC3, GSU, MKN1, MKN7, MKN28, MKN45 and MKN74, were cultured in RPMI-1640 medium (Sigma), supplemented with penicillin, streptomycin and 10% fetal bovine serum, in plastic culture dishes (Corning). Spheres were cultured in Gibco® Dulbecco’s modified Eagle’s medium with nutrient mixture F-12 (Invitrogen), supplemented with 20 ng/ml human recombinant epidermal growth factor (Promega), 20 ng/ml basic fibroblast growth factor (PeproTech Inc.), B-27® Supplement (Invitrogen) and N2 Supplement (Wako), in low-attachment dishes (Corning).

Cell staining and flow cytometry

Cultured cells were harvested and stained using an Aldefluor® stem cell detection kit (StemCell Technologies) for 45 min at 37°C. To stain cell surface markers, cells were incubated on ice with antibodies against CD44, CD26, CD117, CD90 (all from BD Biosciences), EpCAM (BioLegend) and CD133 (Miltenyi). Isotype antibodies were used as the negative controls. Discrimination between live and dead cells was carried out using the Live/Dead® Fixable Yellow Dead Cell Stain kit (Invitrogen). Mouse cells were identified by anti-H2kd (eBioscience) and anti-mouse CD45 (eBioscience) antibodies.

Primary surgical specimens and xenografts

Tumor tissues were digested into single cells with collagenase (Roche) and DNase (Worthington) at 37°C for 1 h. Staining for fluorescence-activated cell sorting (FACS) analysis was performed, as described above. For xenografting, cells were injected subcutaneously with Matrigel® into NOD/SCID mice. All the animal experiments were performed with approval of Animal Experiments Committee of Osaka University.

RNA extraction, cDNA synthesis and quantitative PCR

Total RNA was extracted using TRIzol® reagent. cDNA was synthesized using SuperScript® (Invitrogen). Quantitative PCR was performed using LightCycler® 480 Real-Time PCR system. All procedures were performed according to the manufacturer’s instructions.

Statistical analysis

Statistical significance was determined using the Student’s t-test. Analyses were performed using JMP software.

Results

Screening of CSC markers in GC cell lines

We examined novel markers in gastric CSCs, if: i) they had been previously reported in other tumor types and for which useful antibodies were available for FACS analysis; ii) they were expressed in small populations (<50%) in the cell lines; and iii) these observations were evident in more than half the cell lines.

We investigated the functions of cell surface markers and intracellular molecules (ALDH) to establish a functional detection system. We screened six GC cell lines (AGS, NUGC3, GSU, MKN7, MKN1 and MKN45) for gastric CSC markers using eight candidate markers expressed in other CSCs (CD13, CD26, CD44, CD90, CD117, CD133, EpCAM and ALDH) (11). As shown in Table I, FACS analysis revealed a high expression of EpCAM in all the cell lines (almost 100% postive cells), whereas CD90, CD117 and CD133 expression was uniformly undetectable or negative. CD26 expression was positive (>50%) in four of the cell lines, but undetectable or negative in the other two, suggesting that CD26 expression depends on individual cell lines rather than the heterogeneous conditions of cell line subpopulations. CD13 expression was detected in only one cell line, GSU. Conversely, the investigation of ALDH indicated that the proportion of cells highly expressing ALDH (ALDHhigh cells) was relatively small (6.2–45.5%) compared with the other markers (CD13, CD26, CD90, CD117, CD133 and EpCAM; Table I and Fig. 1A). Moreover, the ALDHhigh cell populations reproducibly disappeared upon the addition of the ALDH inhibitor, diethylaminobenzaldehyde (DEAB), indicating the specificity of detection in ALDHhigh cell populations. Reportedly, ALDH1A1, a substrate for DEAB inhibition, has been shown to be responsible for ALDH activity in CSCs (15). Thus, we focused on ALDH activity.

Table I

Screening for common CSC markers using six GC cell lines.

Table I

Screening for common CSC markers using six GC cell lines.

ALDHCD44EpCAMCD133CD13CD26CD90CD117
AGS+++
GSU+++++++++
NUGC3++++
MKN1+++++++
MKN7+++++++
MKN45+++++++

−, <1%.

+, <50%.

++, 50–100%.

Cells expressing high levels of ALDH also express CD44

We examined CD44 expression, reportedly a CSC marker in breast, colon, esophageal and gastric cancers (11,13,14). Two-dimensional analysis data indicated that ALDHhigh cell populations represented only a small subpopulation of CD44-positive cells, suggesting that ALDH is a good candidate as a CSC marker in GC (Table 1 and Fig. 1B).

Cells expressing high ALDH exist in xenografts in immunodeficient mice

We then examined the involvement of ALDHhigh cell populations in human primary tumor samples. Primary tumor tissues from surgical specimens were obtained with written informed consent and inoculated into immunodeficient NOD/SCID mice. Approximately 30% of inoculated primary samples formed tumors in the mice after several weeks. The probability of tumor formation is likely influenced by tumor tissue viability (nutrients, necrosis and therapy-related damage), vasculogenesis in the mice (dependent on local conditions) and CSC conditions within primary samples. The tumor sample from a patient was subjected to FACS analysis. The data indicated that 57% of the human living tumor cells (separated by FACS using the Live/Dead Fixable Yellow Dead Cell Stain system and distinguished from mouse cells using anti-H2kd and CD45 antibodies) expressed active ALDH (Fig. 1C), indicating that ALDHhigh cells are present in primary human tumor sample.

Sphere formation and tumorigenicity of populations expressing high levels of ALDH

We then examined stemness in ALDHhigh cells. A culture of FACS-sorted ALDHhigh cells in serum-free medium resulted in the frequent formation of large spheres compared with cells expressing low ALDH (ALDHlow cells; Fig. 2A and B), suggesting that ALDHhigh cells possess a greater self-renewal ability, a critical characteristic of CSCs (2,11). We examined tumorigenicity in vivo by inoculating FACS-sorted ALDHhigh and ALDHlow MKN45 cells subcutaneously into NOD/SCID mice. We performed a limiting dilution experiment by reducing the number of inoculating cells. The inoculation of 500 ALDHhigh, but not ALDHlow cells resulted in tumor formation in three out of four mice (Fig. 2C). The inoculation of 5,000 cells resulted in tumors being formed from the ALDHhigh and ALDHlow cells (Fig. 2C). Taken together, these observations indicate that, although multiple factors may be involved, ALDH function is closely associated with the initiation, maintenance and progression of CSCs in vitro and in vivo.

Chemoresistance in cells highly expressing ALDH and the underlying mechanisms

Our aim was the identification of novel molecular therapeutic targets. Reportedly, CSCs can survive toxic injuries and chemoradiation therapy (2,10,11). To combat this, we explored the effect of chemotherapeutic agents commonly used to treat GC. The exposure of the cell cultures to cisplatin and 5-fluorouracil (5-FU) increased the number of ALDHhigh cells (3.1–4.4% with cisplatin and 31.0% with 5-FU treatment; Fig. 3A), indicating that exposure to these chemotherapeutic agents causes an accumulation of surviving ALDHhigh CSCs. To elucidate the molecular mechanisms underlying this chemoresistance, we examined gene expression changes in candidate pathways (11). We found that Notch1 and Sonic hedgehog (Shh) expression was increased in ALDHhigh, compared to ALDHlow cells, suggesting that the survival of ALDHhigh cells following chemotherapy is associated with increased Notch1 and Shh signaling.

Discussion

The demonstration of an association between ALDH and tumors was first shown in breast cancer (16) and subsequently in pancreatic (17), liver (18), colorectal (19), head and neck (19), thyroid (20) and lung (21) cancers. In lung cancer, ALDH activity has been shown to be selective for adenocarcinoma stem cells, depending on Notch signaling (21), in agreement with our observations of gastric CSCs. We demonstrated that in GC, Notch and Shh signaling may be important for both CSC maintenance and the generation of chemoresistance, providing the rationale for further study of therapy-resistant ALDHhigh CSCs. ALDH is widely used as a marker to identify and isolate various types of normal stem cells and CSCs (22). In GC, several markers reportedly characterize CSCs: CD133 (23), CD44 (2326), side-populations identified by FACS (27), CD44 and EpCAM (25), CD54 (26) and CD90 (28). Of these markers, CD44 and ALDH are involved in aerobic glycolysis during cancer metabolism. Although an association between ALDH and the clinicopathlogical features of GC has been reported (29), the relevance of ALDH to chemoresistance has yet to be fully investigated; another study detected no association between immunohistochemical staining for ALDH and prognosis in GC patients (23). In this study, we examined for the first time the involvement of ALDH in chemoresistance and identified a candidate underlying molecular mechanism for this resistance.

GC is the second major cause of cancer-related mortality worldwide and is prevalent across Asia. Helicobacter pylori (H. pylori) infection was identified in 1982 by Marshall and Warren in patients with chronic gastritis and gastric ulcers (30). H. pylori-associated GC has been investigated in order to elucidate the mechanisms underlying gastric tissue damage. In general, the two mechanisms by which H. pylori promotes cancer are as follows: i) enhanced production of free radicals proximal to the H. pylori infection site, increasing the host cell mutation rate; and ii) pregenetic factors that transform host cell phenotypes by altering adhesion proteins or inflammation-related cytokines/chemokines, such as tumor necrosis factor-α or interleukin-6. Thus, H. pylori infection causes enhanced migration or invasion of damaged epithelial cells, without additional tumor suppressor gene mutations (31). Those non-cell autonomous mechanisms are likely facilitated by the hypoxic microenvironment of tumors, since recent studies have implicated hypoxia in inflammatory reactions provoked by H. pylori infection (32). Indeed, hypoxia-inducible factor-1α is mediated by the induction of a ROS-inducible protein (apurinic/apyrimidinic endonuclease 1) and its enhanced interaction with the transcriptional coactivator, p300, leads to transformed phenotypes in H. pylori-infected gastric epithelia (33). Although H. pylori infection and related atrophic gastritis are closely associated with GC, hypoxia and its related metabolism play a critical role in tumor initiation and progression in the stomach and likely in other organs (34). Further studies are warranted to elucidate the association between H. pylori infection and ALDH-positive CSCs in hypoxic areas and to evaluate the eradication of H. pylori infection and GC treatment by surgery, chemotherapy and molecular targeting of therapy-resistant CSC functions.

Acknowledgements

We thank Miyuki Ozaki and Yuko Noguchi for technical support. The current study was partly supported by a Core Research Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, Japan (to H.I. and M.M.); a Grant-in-Aid from the Third Term Comprehensive 10-year Strategy for Cancer Control of the Ministry of Health, Labour and Welfare, Japan (to H.I. and M.M.); a grant from the Kobayashi Cancer Research Foundation (to H.I.); a grant from the Princess Takamatsu Cancer Research Fund, Japan (to H.I.); and a grant from the SENSHIN Medical Research Foundation (to H.I.).

References

1 

Bonnet D and Dick JE: Human acute leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 3:730–737. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Reya T, Morrison SJ, Clarke MF and Weissman IL: Stem cells, cancer and cancer stem cells. Nature. 414:105–111. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Prince ME, Sivanandan R, Kaczorowski A, et al: Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA. 104:973–978. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Haraguchi N, Utsunomiya T, Inoue H, Tanaka F, Mimori K, Barnard GF and Mori M: Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cell. 24:506–513. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C and De Maria R: Identification and expansion of human colon-cancer-initiating cells. Nature. 445:111–115. 2007. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ and Clarke MF: Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 100:3983–3988. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Piccirillo SG, Reynolds BA, Zanetti N, et al: Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature. 444:761–765. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Bao S, Wu Q, McLendon RE, et al: Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 444:756–760. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Ishii H, Iwatsuki M, Ieta K, Ohta D, Haraguchi N, Mimori K and Mori M: Cancer stem cells and chemoradiation resistance. Cancer Sci. 99:1871–1877. 2008. View Article : Google Scholar

11 

Dewi DL, Ishii H, Kano Y, et al: Cancer stem cell theory in gastrointestinal malignancies: recent progress and upcoming challenges. J Gastroenterol. 46:1145–1157. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Haraguchi N, Ishii H, Mimori K, et al: CD13 is a therapeutic target in human liver cancer stem cells. J Clin Invest. 120:3326–3339. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Tamada M, Nagano O, Tateyama S, et al: Modulation of glucose metabolism by CD44 contributes to antioxidant status and drug resistance in cancer cells. Cancer Res. 72:1438–1448. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Ishimoto T, Nagano O, Yae T, et al: CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc(−) and thereby promotes tumor growth. Cancer Cell. 19:387–400. 2011.PubMed/NCBI

15 

Marcato P, Dean CA, Giacomantonio CA and Lee PW: Aldehyde dehydrogenase: its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle. 10:1378–1384. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Ginestier C, Hur MH, Charafe-Jauffret E, et al: ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 1:555–567. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Feldmann G, Dhara S, Fendrich V, et al: Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 67:2187–2196. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Ma S, Chan KW, Lee TK, Tang KH, Wo JY, Zheng BJ and Guan XY: Aldehyde dehydrogenase discriminates the CD133 liver cancer stem cell populations. Mol Cancer Res. 6:1146–1153. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Huang EH, Hynes MJ, Zhang T, et al: Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Res. 69:3382–3389. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Todaro M, Iovino F, Eterno V, et al: Tumorigenic and metastatic activity of human thyroid cancer stem cells. Cancer Res. 70:8874–8885. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Sullivan JP, Spinola M, Dodge M, et al: Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res. 70:9937–9948. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Ma I and Allan AL: The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Rev. 7:292–306. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Wakamatsu Y, Sakamoto N, Oo HZ, et al: Expression of cancer stem cell markers ALDH1, CD44 and CD133 in primary tumor and lymph node metastasis of gastric cancer. Pathol Int. 62:112–119. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Takaishi S, Okumura T, Tu S, et al: Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cell. 27:1006–1020. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Han ME, Jeon TY, Hwang SH, et al: Cancer spheres from gastric cancer patients provide an ideal model system for cancer stem cell research. Cell Mol Life Sci. 68:3589–3605. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Chen T, Yang K, Yu J, et al: Identification and expansion of cancer stem cells in tumor tissues and peripheral blood derived from gastric adenocarcinoma patients. Cell Res. 22:248–258. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Fukuda K, Saikawa Y, Ohashi M, et al: Tumor initiating potential of side population cells in human gastric cancer. Int J Oncol. 34:1201–1207. 2009.PubMed/NCBI

28 

Jiang J, Zhang Y, Chuai S, et al: Trastuzumab (herceptin) targets gastric cancer stem cells characterized by CD90 phenotype. Oncogene. 31:671–682. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Katsuno Y, Ehata S, Yashiro M, Yanagihara K, Hirakawa K and Miyazono K: Coordinated expression of REG4 and aldehyde dehydrogenase 1 regulating tumourigenic capacity of diffuse-type gastric carcinoma-initiating cells is inhibited by TGF-β. J Pathol. 228:391–404. 2012.PubMed/NCBI

30 

Marshall BJ and Warren JR: Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet. 16:1311–1315. 1984. View Article : Google Scholar : PubMed/NCBI

31 

Suganuma M, Yamaguchi K, Ono Y, et al: TNF-alpha-inducing protein, a carcinogenic factor secreted from H. pylori, enters gastric cancer cells. Int J Cancer. 123:117–122. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Sinkovics JG: Molecular biology of oncogenic inflammatory processes. I. Non-oncogenic and oncogenic pathogens, intrinsic inflammatory reactions without pathogens and microRNA/DNA interactions (Review). Int J Oncol. 40:305–349. 2012.

33 

Bhattacharyya A, Chattopadhyay R, Hall EH, Mebrahtu ST, Ernst PB and Crowe SE: Mechanism of hypoxia-inducible factor 1 alpha-mediated Mcl1 regulation in Helicobacter pylori-infected human gastric epithelium. Am J Physiol Gastrointest Liver Physiol. 299:G1177–G1186. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Barker HE, Cox TR and Erler JT: The rationale for targeting the LOX family in cancer. Nat Rev Cancer. 12:540–552. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 42 Issue 4

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
Nishikawa S, Konno M, Hamabe A, Hasegawa S, Kano Y, Ohta K, Fukusumi T, Sakai D, Kudo T, Haraguchi N, Haraguchi N, et al: Aldehyde dehydrogenasehigh gastric cancer stem cells are resistant to chemotherapy. Int J Oncol 42: 1437-1442, 2013
APA
Nishikawa, S., Konno, M., Hamabe, A., Hasegawa, S., Kano, Y., Ohta, K. ... Ishii, H. (2013). Aldehyde dehydrogenasehigh gastric cancer stem cells are resistant to chemotherapy. International Journal of Oncology, 42, 1437-1442. https://doi.org/10.3892/ijo.2013.1837
MLA
Nishikawa, S., Konno, M., Hamabe, A., Hasegawa, S., Kano, Y., Ohta, K., Fukusumi, T., Sakai, D., Kudo, T., Haraguchi, N., Satoh, T., Takiguchi, S., Mori, M., Doki, Y., Ishii, H."Aldehyde dehydrogenasehigh gastric cancer stem cells are resistant to chemotherapy". International Journal of Oncology 42.4 (2013): 1437-1442.
Chicago
Nishikawa, S., Konno, M., Hamabe, A., Hasegawa, S., Kano, Y., Ohta, K., Fukusumi, T., Sakai, D., Kudo, T., Haraguchi, N., Satoh, T., Takiguchi, S., Mori, M., Doki, Y., Ishii, H."Aldehyde dehydrogenasehigh gastric cancer stem cells are resistant to chemotherapy". International Journal of Oncology 42, no. 4 (2013): 1437-1442. https://doi.org/10.3892/ijo.2013.1837