Minireviews Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Nov 26, 2014; 6(5): 606-613
Published online Nov 26, 2014. doi: 10.4252/wjsc.v6.i5.606
Stem cells in gastrointestinal cancers: The road less travelled
Sameh Mikhail, the Ohio State University, Comprehensive Cancer Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, United States
Amer Zeidan, Yale Comprehensive Cancer Center, Section of Hematology, Department of Internal Medicine, Yale University, New Haven, CT 06520-8028, United States
Author contributions: Mikhail S and Zeidan A contributed to this paper.
Correspondence to: Amer Zeidan, MBBS, MhS, Yale Comprehensive Cancer Center, Section of Hematology, Department of Internal Medicine, Yale University, 333 Cedar street, PO. Box 208028, New Haven, CT 06520-8028, United States. amer.zeidan@yale.edu
Telephone: +1-203-7377103 Fax: +1-203-7857232
Received: July 25, 2014
Revised: August 29, 2014
Accepted: September 6, 2014
Published online: November 26, 2014

Abstract

Cancer stem cells (CSC) are thought to be malignant cells that have the capacity to initiate and maintain tumor growth and survival. Studies have described CSC in various gastrointestinal neoplasms such as colon, pancreas and liver and gastroesophageal tumors. The mechanism by which CSC develop remains unclear. Several studies have explored the role of dysregulation of the Wnt/β-catenin, transformation growth factor-beta and hedhog pathways in generation of CSC. In this review, we discuss the various molecular abnormalities that may be related to formation of CSC in gastrointestinal malignancies, strategies to identify CSC and therapeutic strategies that are based on these concepts. Identification and targeting CSC is an intriguing area and may provide a new therapeutic option for patients with cancer including gastrointestinal malignancies. Although great progress has been made, many issues need to be addressed. Precise targeting of CSC will require precise isolation and characterization of those cells. This field is also evolving but further research is needed to identify markers that are specific for CSC. Although the application of this field has not entered the clinic yet, there continues to be significant optimism about its potential utility in overcoming cancer resistance and curing patients with cancer.

Key Words: Cancer stem cells, CD133+, WNT/β-catenin, Transformation growth factor-beta, Hedgehog, Notch

Core tip: Cancer stem cells (CSC) are thought to be malignant cells that have the capacity to initiate and maintain tumor growth and survival. Several studies have explored the role of dysregulation of the Wnt/β- catenin, transformation growth factor-beta and hedhog pathways in generation of CSC. The exact machismo of their development, however, remains unknown. Several investigators have researched modalities to identify and target CSC. In this review, we summarize the recent evidence exploring the mechanisms of development, identification and targeting of CSC in gastrointestinal malignancies.



STEM CELLS IN GASTROINTESTINAL CANCERS: THE ROAD LESS TRAVELLED

Cancer is a disease of adult stem cells (SC). Adult SC are the only cells that persist in the tissue for a sufficient length of time to acquire the sufficient sequential genetic alterations for cancer development[1]. Adult SC have been traditionally relatively quiescent, a feature thought to protect them from the accumulation of DNA errors that may lead to carcinogenesis[1]. In the gastrointestinal tract, the immediate stem cell progeny, however, proliferate rapidly to allow for tissue repopulation[1]. Their limited life span restricts the impact of any replication errors. It is worth noting that this concept has been challenged by recent studies that suggest that adult stem cells are in fact capable of rapid self-renewal[2]. Similarly, cancer stem cells (CSC) have the capacity to initiate and maintain tumor growth and survival[3]. Studies have described CSC in gastrointestinal neoplasms such as colon, pancreas and liver[4-6]. The mechanism by which CSC develop remains unclear[1]. Several studies have explored the role of dysregulation of the Wnt/β-catenin, transformation growth factor-beta (TGF-β) and hedhog pathways in generation of CSC[7-9]. In this review, we discuss the various molecular abnormalities that may be related to formation of CSC in gastrointestinal malignancies, strategies to identify CSC and therapeutic strategies that are based on these concepts.

MOLECULAR PATHWAYS ASSOCIATED WITH CSCS IN GASTROINTESTINAL MALIGNANCIES
Notch signaling pathway

The Notch signaling pathway plays an important role in embryogenesis, cellular homeostasis-, differentiation and apoptosis[10-12]. While Notch mediates a number of biological processes through the “canonical “Notch signaling pathway, it also mediates a ligand- or transcription independent function known as the “non-canonical” pathway[12,13]. The canonical Notch pathway includes at least four Notch receptors (Notch 1-4) and five Notch ligands Delta-like 1,3 and 4 and Jagged 1 and 2[14]. When Notch ligand binds to a Notch receptor, Notch will be cleaved through a series of proteolytic cleavages by multiple enzymes leading to release of the active Notch fragment and activation of Notch target genes[15]. Notch target genes include Akt, mTOR (mammalian target of rapamycin, NF-κB, c-Myc and VEGF (vascular endothelial growth factor) and cyclin D1[16,17]. Activation of the Notch pathway can have tumor suppressor function in HCC but may play on oncogenic role in colon and pancreatic cancers[14]. Notch signaling has been found to play a pivotal role in CSC. Overexpression of Notch-1 and -2 was observed in pancreatic CSC and was associated with increased expression of CSC surface markers such as CD44 and EpCAM[15,17-19]. This observation suggests that Notch signaling may be involved in pancreatic CSC self-renewal but will need further confirmation.

WNT/β-catenin pathway

Notch signaling also perform a “non-canonical role” through antagonizing Wnt/β-catenin signaling[12,13]. Disrupted Wnt signaling is observed in a variety of gastrointestinal cancers which underscores its importance in carcinogenesis[20]. The Wnt pathway plays a crucial role in embryogenesis with signaling effects that regulate proliferation and apoptosis in developing cells[21]. Wnt pathway activation plays a fundamental role in maintenance of SC compartment and regulation of cellular differentiation[22]. The “canonical” Wnt pathway plays a crucial role in modulating the balance between self-renewal and differentiation in several adult CSC[21]. The “canonical” Wnt pathway describes a sequence of events beginning with the translocation of β-catenin from the cell membrane into the nuclear, where β-catenin then acts as a co-activator of the TCF/LEF family of transcription factors[23,24]. The signaling cascade is typically initiated when Wnt ligand binds to Frizzled (FZD), a transmembrane receptor[23]. The transcription factors activated by β-catenin subsequently regulate specific target genes including c-myc, cyclin D1 and survivin. FZD binding to Wnt ligand also promotes the escape of β-catenin from its association with E-cadherin[23,25]. The cytoplasmic elements of the activated Wnt pathway prevent β-catenin from being phosphorylated by degradation complex composed of a serine-threonine kinase, glycogen synthase kinase-3β (GSK3β), protein scaffolds, AXIN and adenomatosis polyposis coli (APC)[25]. Mutations of these proteins allow β-catenin to accumulate in the nucleus to enhance the transcription of its target genes which are found in many cancers[9]. For example, in hepatocellular carcinoma (HCC), mutations of β-catenin are located in exon 3 of CTNNB1 gene which is the phosphorylation site for GSK3B, AXIN1 and AXIN2 mutation[26]. It is worth noting that 20%-40% of human HCC exhibit abnormal cytoplasmic and nuclear accumulation of β-catenin by immunohistochemistry (IHC)[27]. Β-Catenin can also undergo downregulation via the non-canonical Notch pathway. In this case, membrane-bound Notch forms a complex with active Β-Catenin in the presence of Wnts. This action degrades active Β-Catenin and thus inhibits its pathway. This process allows for regulation of SC and its dysfunction could lead to expansion of CSC[13]. Markers for elevated expression of Wnt include CD133+ and EpCAM+[28]. The knockdown of expression of EpCAM, in HCC stem cells resulted in decreased proliferation, colony formation, migration and drug resistance which highlight the role and Wnt signaling in tumor survival[28,29]. Additionally, knockdown of β-catenin resulted in inhibition of CSC[30]. Similarly mutations in APC gene acts to suppress Wnt signaling and result in familial adenomatous polyposis (FAP) syndrome[31]. In the majority of sporadic colorectal cancers, loss of APC or β-catenin mutations seems to be early events in carcinogenesis[32]. Of note, Apc 1638N has been shown to result in multiple intestinal tumors in mice[32].

TGF-βpathway

TGF-β signaling is crucial for self-renewal and maintenance of SC and in the formation of gastrointestinal cancers[8,33]. TGF-β forms a complex with the serine-threonine kinase receptor type I and II[34]. The receptors are activated sequentially and subsequently phosphorylate one of the receptor-activated R-mads[35]. The activated R-mad will heterodimerize with Smad4 and then translocate to the nuclear to regulate gene transcription[36]. Disruption of TGF-β signaling results in dysregulated gene expression and hence gastrointestinal malignancies are associated with suppressed activity of different members of TGF-β pathway[37,38]. For example, inactivation of Smad4 is seen in approximately 50% of patients with pancreatic cancer[39]. Similarly, reduced Smad4 expression and loss of ELF, a modulator of activity of Smad3, are observed in human colon and gastric cancer tissue[40,41]. Additionally, inactivating mutation of TGF-β II receptor was described in colon cancer[37].

Hedgehog pathway

The Hedgehog signaling pathway consists of a complex of molecules which regulate cell differentiation, regeneration and stem cell biology[9]. The pathway plays a central role in the development and homeostasis of the gut tissue[9]. The Hedgehog pathway is deregulated in gastrointestinal cancers[42]. Up to 60% of HCC samples express Sonic, the predominant ligand of the hedgehog pathway[42]. Additionally, genes involved in the hedgehog pathway are highly expressed in CD133+ liver cancer SC[43]. It is worth noting that suppression of Hedgehog pathway decreased HCC cell proliferation and sensitized HCC cells to treatment with 5-fluorouracil[44]. Hedgehog signaling has been shown to be essential for proliferation and survival of human colon cancers[45]. It is thought to affect both tumor growth and CD133+ CSC[45]. Similarly, HH signaling has been associated with pancreas cancer invasion and metastasis. Conversely inhibition of HH signaling inhibited pancreatic metastatic spread[46].

PTEN pathway

PTEN is a phosphatase that antagonizes PI3 kinase activity[47]. PTEN helps control the proliferative rate and the number of intestinal stem cells and its loss is associated with an increase in intestinal SC[47]. It is also thought that PTEN pathway controls SC activation via interaction with the Wnt pathway[48]. It is also proposed that PTEN pathway interacts with the TGF-β pathway described above[48]. Mutations in PTEN, result in a cancer syndrome (Cowden’s syndrome) characterized by hamartomas in the gastrointestinal tract, central nervous system and skin in addition to tumors in the breast and thyroid gland[49]. PTEN deficient mice exhibit increase in intestinal SC which results in excess crypt formation[47].

Identification of CSCs

Eradication of CSC stems is an intriguing concept that provides hope in the possibility of finding a cure for cancer. Any therapeutic modality that targets CSC will require accurate identification and characterization of the CSC and differentiating them from normal SC. Isolation of cancer cells through the identification of pathognomonic surface markers has recently gained popularity and is an area of active investigation[50,51]. CD133+ emerged as a promising surface marker for CSC[50]. Singh et al[51] used flow cytometry to successfully isolate CD133+ CSC in human brain tumors and implanted them into forebrain of immunodeficient mice. Transplantation of as few as 100 cells produced tumors that were phenotypically similar to original tumors. Similar findings were reported in colorectal cancer. Several groups isolated subpopulations of cells, accounting for approximately 1% of total number of cells within a tumor, that were CD133+ and we capable initiating cancer when transplanted in immunodeficient mice[5,52,53]. Other studies have identified new CSC markers (Table 1) that may be promising in isolation of CSC such as Lgr5, CD44, CD24 and epithelial specific antigen[54-57]. These markers were isolated in HCC and pancreatic cancer. This field is currently in evolution. Efforts have been made to identify surface marker “signatures “ that are specific for each type of cancer (Table 2) It is worth noting that isolation of cancer cells is far from perfect and remains an area of controversy. Not all CSC express SC markers and some tumor cells that are not SC may also express those markers[1]. Great progress has been already made in this area but this more works remains to be done.

Table 1 Markers used in gastrointestinal cancer stem cell identification.
MarkersRef.
CD133+[34]
CD44+[55]
CD24+[84]
Lgr5[53]
mTert[85]
Olfm4[86]
Ascl2[87]
ALDH[79]
Sox9[88]
Msi 1[89]
Dcamkl1[90]
Table 2 Surface markers of gastrointestinal cancer stem cell.
Tumor typePhenotype of CSC markersRef.
LiverCD133+, CD49f+, CD90+[1,6,91]
ColonCD133+, CD44+, CD166+, EpCAM+, CD24+[5,45,52]
PancreaticCD133+, CD44+, EpCAM+, CD24+[57]
StomachCD44+, CD133+, NESTIN, CD90+, CD54+, ALDH1[79]
Resistance of CSCs to anticancer therapy

Several studies demonstrated that CSC exhibit resistance to chemotherapy agent[2,58]. One of the widely accepted theories is that the elevated levels of ATP-binding cassette (ABC) transporters mediate resistance to chemotherapy[2,3,58,59]. ATP transporters are membrane transporters that can pump small molecules including cytotoxic drugs out of cells in exchange for ATP hydrolysis[59]. CSC as well as normal SC appear to express high levels of ABC transporters[60]. This characteristic can lead to multidrug resistance and enhanced tumorigenesis. Evolving evidence suggests that numerous cell lines and tumors contain CSC, referred to as side population (SP) cells that possess a differentially greater capacity to resist chemotherapeutic agents and invade surrounding tissues[2,61-63]. This phenomenon, however, may allow for development of therapies that could target ATP transporters in CSC.

Targeting CSCs

Targeting CSC is an intriguing concept that may offer several therapeutic advantages. Targeting the inherently resistant CSC may overcome resistant to chemotherapeutic agents. Most patients with metastatic gastrointestinal cancers tend to experience treatment failure and resistance to palliative chemotherapy[64-66]. Additionally, targeting CSC may, not only improve efficacy of treatment but may also reduce therapy-related toxicity through developing treatment that are selective for CSC and not toxic to healthy tissues. Novel treatment strategies are, therefore, being developed that target surface markers on CSC, ATP-binding cassettes, key signaling pathways or their tumor microenvironment[1].

Targeting surface markers: Since CD133+ is expressed in CSC in gastrointestinal cancer, it represents an interesting target to selectively inhibit CSC. A recent study demonstrated that carbon nanotubes conjugated with CD133+ monoclonal antibodies caused photothermolysis of CD133+ glioblastoma cells when affixed to an anti-CD133 antibody that selectively targeted those cells[67]. This study represents an encouraging proof of concept that gastrointestinal CSC can be possibly targeted with similar strategies.

Targeting cancer stem cell pathways: Targeting signaling pathways that are thought to be active in CSC is an ongoing area of active research. Lin et al demonstrated that a curcumin analogue, GO Y030, may have clinical activity against colorectal cancer SC in vitro and vivo[68]. They identified aldehydehehydrogenase (ALDH) positive and CD133+ colorectal CSC using flow cytometry. The demonstrated that isolated CSC exhibited STAT-3 (signal transducers and activators of transcription-3) activation and treated them with GO-Y030. GO-Y030 inhibited STAT3 phosphorylation and reduced STAT3 downstream target gene expression resulting in induction of apoptosis in colon CSC. Additionally, GO-Y030 suppressed tumor and CSC growth of SW480 and HCT-116 colon cancer cell lines in vivo in mouse models. Interestingly, Curcumin has been shown to also inhibit cell growth and apoptosis in pancreatic cancer cells. Its effect was associated with down-regulation of Notch-1 expression, which suggests that Curcumin may be associated with potential advantageous activity against pathways that are upregulated in CSC[18]. Other attempts to target Notch signaling in gastrointestinal CSC have, however, not been very successful. Gamma-secretase inhibitors (GSI) are thought to antagonize Notch signaling through blocking of Notch receptor cleavage[69]. Evaluation of the effect of GSI in two gastric cancer cell lines did not result in any appreciable anti-tumor effects[70]. These results were surprising since GSI have shown promising antitumor potential in leukemia, breast and glioblastoma multiformes models[71-73].

Evolving evidence suggests that targeting the Hedgehog pathway may be a feasible strategy to inhibit CSC. Cyclopamine, a naturally occurring hedgehog inhibitor has shown promising potential[46]. As a single agent cyclopamine suppressed the invasion of pancreatic cancer cells[4]. Cyclopamine reduced the percentage of cells expressing the pancreatic CSC markers such as ALDH[74]. In combination with gemcitabine, cyclopamine resulted in reduction of metastasis in an orthotopic xenograft model[74]. To further clarify this observation, Yao et al[74] demonstrated that cyclopamine dowregulated the expression of CD44 and CD133+ in gemcitabine-resistant pancreatic cancer cells indicating that it may be an effective modality for reversing gemcitabine resistance in pancreatic CSC. A similar observation was made in gastric CSC where blocking of Hedgehog pathway with cyclopamine decreased self-renewing properties and enhanced sensitivity of gastric cancer cells to chemotherapeutic agents[75]. Additionally, Feldmann et al[76] demonstrated that IPI-269609, a novel Hedgehog inhibitor, inhibited growth and metastasis of pancreatic cancer mostly through targeting of the CSC.

Since the Wnt pathway is also deregulated in CSC, it represents an intriguing target for cancer treatment. Anti-Wnt therapy is in early stages of clinical development[77]. He et al[77] demonstrated that a monoclonal antibody against Wnt-1 induced apoptosis in human cancer cells. Also, Salinomycin, an antibiotic commonly used in poultry firmly, is thought to suppress Wnt/β-catenin signal transduction[78]. In gastric cancer, salinomycin, selectively inhibited gastric CSC in vitro[79]. Wnt inhibitors also are being investigated in phase I clinical trials. Oral LGK974[80] is a potent and specific inhibitor of O-acyltransferase Porcupine (Porcn) that acetylates Wnt proteins required for their biological activities is being investigated in a phase I clinical trial in patients with malignancies dependent on Wnt ligands. This trial is enrolling patients with pancreatic and colon adenocarcinoma.

Targeting ATP-driven efflux transporters has been explored in preclinical and early phase clinical trials. The first drug efflux pump inhibitor is verapamil. Simultaneous treatment with verapamil and chemotherapy resulted in promising antitumor activity. Other agents such as Dofequidar Fumarate (MS-209), Biricolar (VX-710), and tariquidar are in various stages of clinical development[81-83]. Most of the experience with these agents is derived from lung and breast cancer trials but these agents, to our knowledge, have not been investigated in gastrointestinal cancers.

CONCLUSION

Identification and targeting CSC is an intriguing area and may provide a new therapeutic option for patients with cancer including gastrointestinal malignancies. It is a rapidly evolving area in the treatment of gastrointestinal and other tumors. Although great progress has been made, many issues need to be addressed. The CSC model does not fully explain the observed genetic heterogeneity of many tumors. This criticism may however be explained by the fact that even CSC may evolve over time and give rise to cells that are both genetically and functionally heterogeneous[1]. Furthermore, accurate targeting of CSC will require precise isolation and characterization of those cells. This field is also evolving but further research is needed to identify markers that are specific for CSC. Nevertheless, there continues to be significant excitement about this field and hope that it may represent a new treatment modality in patients with cancer.

Footnotes

P- Reviewer: Han X, Kamer E, Pan WS S- Editor: Song XX L- Editor: A E- Editor: Lu YJ

References
1.  Chen K, Huang YH, Chen JL. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin. 2013;34:732-740.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 433]  [Cited by in F6Publishing: 411]  [Article Influence: 37.4]  [Reference Citation Analysis (0)]
2.  Ho MM, Ng AV, Lam S, Hung JY. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 2007;67:4827-4833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 702]  [Cited by in F6Publishing: 760]  [Article Influence: 44.7]  [Reference Citation Analysis (0)]
3.  Kruger JA, Kaplan CD, Luo Y, Zhou H, Markowitz D, Xiang R, Reisfeld RA. Characterization of stem cell-like cancer cells in immune-competent mice. Blood. 2006;108:3906-3912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
4.  Jimeno A, Feldmann G, Suárez-Gauthier A, Rasheed Z, Solomon A, Zou GM, Rubio-Viqueira B, García-García E, López-Ríos F, Matsui W. A direct pancreatic cancer xenograft model as a platform for cancer stem cell therapeutic development. Mol Cancer Ther. 2009;8:310-314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 211]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
5.  O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106-110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2977]  [Cited by in F6Publishing: 2949]  [Article Influence: 163.8]  [Reference Citation Analysis (0)]
6.  Alison MR. Characterization of the differentiation capacity of rat-derived hepatic stem cells. Semin Liver Dis. 2003;23:325-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 31]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
7.  Janssens N, Janicot M, Perera T. The Wnt-dependent signaling pathways as target in oncology drug discovery. Invest New Drugs. 2006;24:263-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 44]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
8.  Massagué J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, and heritable disorders. Cell. 2000;103:295-309.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Taipale J, Beachy PA. The Hedgehog and Wnt signalling pathways in cancer. Nature. 2001;411:349-354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1040]  [Cited by in F6Publishing: 1017]  [Article Influence: 44.2]  [Reference Citation Analysis (0)]
10.  Benedito R, Rocha SF, Woeste M, Zamykal M, Radtke F, Casanovas O, Duarte A, Pytowski B, Adams RH. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF-VEGFR2 signalling. Nature. 2012;484:110-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 280]  [Article Influence: 23.3]  [Reference Citation Analysis (0)]
11.  Radtke F, Raj K. The role of Notch in tumorigenesis: oncogene or tumour suppressor? Nat Rev Cancer. 2003;3:756-767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 600]  [Cited by in F6Publishing: 596]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
12.  Ranganathan P, Weaver KL, Capobianco AJ. Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer. 2011;11:338-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 594]  [Cited by in F6Publishing: 607]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
13.  Andersen P, Uosaki H, Shenje LT, Kwon C. Non-canonical Notch signaling: emerging role and mechanism. Trends Cell Biol. 2012;22:257-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 161]  [Cited by in F6Publishing: 163]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
14.  Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it’s NOTCH what you think. J Exp Med. 2011;208:1931-1935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 254]  [Cited by in F6Publishing: 276]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
15.  Bolós V, Blanco M, Medina V, Aparicio G, Díaz-Prado S, Grande E. Notch signalling in cancer stem cells. Clin Transl Oncol. 2009;11:11-19.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Wang Z, Banerjee S, Li Y, Rahman KM, Zhang Y, Sarkar FH. Down-regulation of notch-1 inhibits invasion by inactivation of nuclear factor-kappaB, vascular endothelial growth factor, and matrix metalloproteinase-9 in pancreatic cancer cells. Cancer Res. 2006;66:2778-2784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 241]  [Cited by in F6Publishing: 247]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
17.  Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. Inhibition of nuclear factor kappab activity by genistein is mediated via Notch-1 signaling pathway in pancreatic cancer cells. Int J Cancer. 2006;118:1930-1936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 108]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
18.  Wang Z, Zhang Y, Banerjee S, Li Y, Sarkar FH. Notch-1 down-regulation by curcumin is associated with the inhibition of cell growth and the induction of apoptosis in pancreatic cancer cells. Cancer. 2006;106:2503-2513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 214]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
19.  Wang Z, Zhang Y, Li Y, Banerjee S, Liao J, Sarkar FH. Down-regulation of Notch-1 contributes to cell growth inhibition and apoptosis in pancreatic cancer cells. Mol Cancer Ther. 2006;5:483-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 244]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
20.  Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, Hérault A, Saric J, Belghiti J, Franco D. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology. 2007;45:42-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 827]  [Cited by in F6Publishing: 848]  [Article Influence: 49.9]  [Reference Citation Analysis (0)]
21.  Li Y, Welm B, Podsypanina K, Huang S, Chamorro M, Zhang X, Rowlands T, Egeblad M, Cowin P, Werb Z. Evidence that transgenes encoding components of the Wnt signaling pathway preferentially induce mammary cancers from progenitor cells. Proc Natl Acad Sci USA. 2003;100:15853-15858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 426]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
22.  Veeman MT, Axelrod JD, Moon RT. A second canon. Functions and mechanisms of beta-catenin-independent Wnt signaling. Dev cell. 2003;5:367-77.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Huber AH, Weis WI. The structure of the beta-catenin/E-cadherin complex and the molecular basis of diverse ligand recognition by beta-catenin. Cell. 2001;105:391-402.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Pinto D, Gregorieff A, Begthel H, Clevers H. Canonical Wnt signals are essential for homeostasis of the intestinal epithelium. Genes Dev. 2003;17:1709-1713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 748]  [Article Influence: 35.6]  [Reference Citation Analysis (0)]
25.  Giles RH, van Es JH, Clevers H. Caught up in a Wnt storm: Wnt signaling in cancer. Biochim Biophys Acta. 2003;1653:1-24.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Satoh S, Daigo Y, Furukawa Y, Kato T, Miwa N, Nishiwaki T, Kawasoe T, Ishiguro H, Fujita M, Tokino T. AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1. Nat Genet. 2000;24:245-250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 703]  [Cited by in F6Publishing: 697]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
27.  Fujie H, Moriya K, Shintani Y, Tsutsumi T, Takayama T, Makuuchi M, Kimura S, Koike K. Frequent beta-catenin aberration in human hepatocellular carcinoma. Hepatol Res. 2001;20:39-51.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, Jia H, Ye Q, Qin LX, Wauthier E. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136:1012-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 891]  [Cited by in F6Publishing: 905]  [Article Influence: 60.3]  [Reference Citation Analysis (0)]
29.  Yamashita T, Forgues M, Wang W, Kim JW, Ye Q, Jia H, Budhu A, Zanetti KA, Chen Y, Qin LX. EpCAM and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res. 2008;68:1451-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 545]  [Cited by in F6Publishing: 561]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
30.  Teng Y, Wang X, Wang Y, Ma D. Wnt/beta-catenin signaling regulates cancer stem cells in lung cancer A549 cells. Biochem Biophys Res Commun. 2010;392:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 189]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
31.  Haggitt RC, Reid BJ. Hereditary gastrointestinal polyposis syndromes. Am J Surg Pathol. 1986;10:871-87.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Smits R, van der Houven van Oordt W, Luz A, Zurcher C, Jagmohan-Changur S, Breukel C, Khan PM, Fodde R. Apc1638N: a mouse model for familial adenomatous polyposis-associated desmoid tumors and cutaneous cysts. Gastroenterology. 1998;114:275-283.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Chang H, Brown CW, Matzuk MM. Genetic analysis of the mammalian transforming growth factor-beta superfamily. Endocr Rev. 2002;23:787-823.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Tang Y, Kitisin K, Jogunoori W, Li C, Deng CX, Mueller SC, Ressom HW, Rashid A, He AR, Mendelson JS. Progenitor/stem cells give rise to liver cancer due to aberrant TGF-beta and IL-6 signaling. Proc Natl Acad Sci USA. 2008;105:2445-2450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 271]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
35.  Feng XH, Derynck R. Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol. 2005;21:659-693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1427]  [Cited by in F6Publishing: 1459]  [Article Influence: 76.8]  [Reference Citation Analysis (0)]
36.  Mishra L, Derynck R, Mishra B. Transforming growth factor-beta signaling in stem cells and cancer. Science. 2005;310:68-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 156]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
37.  Tang Y, Katuri V, Srinivasan R, Fogt F, Redman R, Anand G, Said A, Fishbein T, Zasloff M, Reddy EP. Transforming growth factor-beta suppresses nonmetastatic colon cancer through Smad4 and adaptor protein ELF at an early stage of tumorigenesis. Cancer Res. 2005;65:4228-4237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 70]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
38.  Katuri V, Tang Y, Marshall B, Rashid A, Jogunoori W, Volpe EA, Sidawy AN, Evans S, Blay J, Gallicano GI. Inactivation of ELF/TGF-beta signaling in human gastrointestinal cancer. Oncogene. 2005;24:8012-8024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Bartsch D, Barth P, Bastian D, Ramaswamy A, Gerdes B, Chaloupka B, Deiss Y, Simon B, Schudy A. Higher frequency of DPC4/Smad4 alterations in pancreatic cancer cell lines than in primary pancreatic adenocarcinomas. Cancer Lett. 1999;139:43-49.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Kim SS, Shetty K, Katuri V, Kitisin K, Baek HJ, Tang Y, Marshall B, Johnson L, Mishra B, Mishra L. TGF-beta signaling pathway inactivation and cell cycle deregulation in the development of gastric cancer: role of the beta-spectrin, ELF. Biochem Biophys Res Commun. 2006;344:1216-1223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
41.  Kitisin K, Ganesan N, Tang Y, Jogunoori W, Volpe EA, Kim SS, Katuri V, Kallakury B, Pishvaian M, Albanese C. Disruption of transforming growth factor-beta signaling through beta-spectrin ELF leads to hepatocellular cancer through cyclin D1 activation. Oncogene. 2007;26:7103-7110.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 101]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
42.  Sicklick JK, Li YX, Jayaraman A, Kannangai R, Qi Y, Vivekanandan P, Ludlow JW, Owzar K, Chen W, Torbenson MS. Dysregulation of the Hedgehog pathway in human hepatocarcinogenesis. Carcinogenesis. 2006;27:748-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 205]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
43.  Huang S, He J, Zhang X, Bian Y, Yang L, Xie G, Zhang K, Tang W, Stelter AA, Wang Q. Activation of the hedgehog pathway in human hepatocellular carcinomas. Carcinogenesis. 2006;27:1334-1340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 175]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
44.  Wang Q, Huang S, Yang L, Zhao L, Yin Y, Liu Z, Chen Z, Zhang H. Down-regulation of Sonic hedgehog signaling pathway activity is involved in 5-fluorouracil-induced apoptosis and motility inhibition in Hep3B cells. Acta Biochim Biophys Sin (Shanghai). 2008;40:819-829.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Roy S, Majumdar AP. Signaling in colon cancer stem cells. J Mol Signal. 2012;7:11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
46.  Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, Mullendore M, Karikari C, Alvarez H, Iacobuzio-Donahue C, Jimeno A. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 515]  [Cited by in F6Publishing: 516]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
47.  He XC, Yin T, Grindley JC, Tian Q, Sato T, Tao WA, Dirisina R, Porter-Westpfahl KS, Hembree M, Johnson T. PTEN-deficient intestinal stem cells initiate intestinal polyposis. Nat Genet. 2007;39:189-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 360]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
48.  Labbé E, Letamendia A, Attisano L. Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways. Proc Natl Acad Sci USA. 2000;97:8358-8363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 342]  [Cited by in F6Publishing: 342]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
49.  Liaw D, Marsh DJ, Li J, Dahia PL, Wang SI, Zheng Z, Bose S, Call KM, Tsou HC, Peacocke M. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet. 1997;16:64-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1451]  [Cited by in F6Publishing: 1340]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
50.  Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire J, Dirks PB. Identification of a cancer stem cell in human brain tumors. Cancer Res. 2003;63:5821-5828.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432:396-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5422]  [Cited by in F6Publishing: 5376]  [Article Influence: 268.8]  [Reference Citation Analysis (0)]
52.  Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445:111-115.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Dalerba P, Dylla SJ, Park IK, Liu R, Wang X, Cho RW, Hoey T, Gurney A, Huang EH, Simeone DM. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci USA. 2007;104:10158-10163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1541]  [Cited by in F6Publishing: 1597]  [Article Influence: 93.9]  [Reference Citation Analysis (0)]
54.  Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003-1007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3854]  [Cited by in F6Publishing: 4011]  [Article Influence: 235.9]  [Reference Citation Analysis (0)]
55.  Durnez A, Verslype C, Nevens F, Fevery J, Aerts R, Pirenne J, Lesaffre E, Libbrecht L, Desmet V, Roskams T. The clinicopathological and prognostic relevance of cytokeratin 7 and 19 expression in hepatocellular carcinoma. A possible progenitor cell origin. Histopathology. 2006;49:138-151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 317]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
56.  Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2377]  [Cited by in F6Publishing: 2352]  [Article Influence: 138.4]  [Reference Citation Analysis (0)]
57.  Gou S, Liu T, Wang C, Yin T, Li K, Yang M, Zhou J. Establishment of clonal colony-forming assay for propagation of pancreatic cancer cells with stem cell properties. Pancreas. 2007;34:429-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 98]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
58.  Xiong B, Ma L, Hu X, Zhang C, Cheng Y. Characterization of side population cells isolated from the colon cancer cell line SW480. Int J Oncol. 2014;45:1175-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
59.  Deeley RG, Westlake C, Cole SP. Transmembrane transport of endo- and xenobiotics by mammalian ATP-binding cassette multidrug resistance proteins. Physiol Rev. 2006;86:849-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 533]  [Article Influence: 29.6]  [Reference Citation Analysis (0)]
60.  Erdei Z, Lőrincz R, Szebényi K, Péntek A, Varga N, Likó I, Várady G, Szakács G, Orbán TI, Sarkadi B. Expression pattern of the human ABC transporters in pluripotent embryonic stem cells and in their derivatives. Cytometry B Clin Cytom. 2014;86:299-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
61.  Matsui W, Wang Q, Barber JP, Brennan S, Smith BD, Borrello I, McNiece I, Lin L, Ambinder RF, Peacock C. Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 2008;68:190-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 394]  [Cited by in F6Publishing: 404]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
62.  Tam SP, Mok L, Chimini G, Vasa M, Deeley RG. ABCA1 mediates high-affinity uptake of 25-hydroxycholesterol by membrane vesicles and rapid efflux of oxysterol by intact cells. Am J Physiol Cell Physiol. 2006;291:C490-C502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
63.  Rothnie A, Callaghan R, Deeley RG, Cole SP. Role of GSH in estrone sulfate binding and translocation by the multidrug resistance protein 1 (MRP1/ABCC1). J Biol Chem. 2006;281:13906-13914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
64.  Benson AB NCCN practive guidelines in Oncology-v.2. 2020;2010-12-11 Available from: https: //urldefense.proofpoint.com/v1/url?u=http: //www.nccn.org/professionals/physician_gls/PDF/hepatobiliary.pdf&k=ux7ohqYFcw1oDo0gOpSLlw== &r=UbmibQFmfYpeICw9U5pTPJzUWGxSLHJd7zX9XiGtctk= &m=pjFYkemH793aikdv8k6btkIbOaLgMln0Tji0H1VIIJk= &s=b2bfd8a0d7cbff133bd164252aedadd205bde5cda51fcc73913ca3054a65aa8b.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Benson AB, Bekaii-Saab T, Chan E, Chen YJ, Choti MA, Cooper HS, Engstrom PF, Enzinger PC, Fakih MG, Fenton MJ. Metastatic colon cancer, version 3.2013: featured updates to the NCCN Guidelines. J Natl Compr Canc Netw. 2013;11:141-152; quiz 152.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Llovet JM, Bruix J. Molecular targeted therapies in hepatocellular carcinoma. Hepatology. 2008;48:1312-1327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 801]  [Cited by in F6Publishing: 840]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
67.  Wang CH, Chiou SH, Chou CP, Chen YC, Huang YJ, Peng CA. Photothermolysis of glioblastoma stem-like cells targeted by carbon nanotubes conjugated with CD133 monoclonal antibody. Nanomedicine. 2011;7:69-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 136]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
68.  Lin L, Liu Y, Li H, Li PK, Fuchs J, Shibata H, Iwabuchi Y, Lin J. Targeting colon cancer stem cells using a new curcumin analogue, GO-Y030. Br J Cancer. 2011;105:212-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 101]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
69.  Grosveld GC. Gamma-secretase inhibitors: Notch so bad. Nat Med. 2009;15:20-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
70.  Sun Y, Gao X, Liu J, Kong QY, Wang XW, Chen XY, Wang Q, Cheng YF, Qu XX, Li H. Differential Notch1 and Notch2 expression and frequent activation of Notch signaling in gastric cancers. Arch Pathol Lab Med. 2011;135:451-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
71.  Tatarek J, Cullion K, Ashworth T, Gerstein R, Aster JC, Kelliher MA. Notch1 inhibition targets the leukemia-initiating cells in a Tal1/Lmo2 mouse model of T-ALL. Blood. 2011;118:1579-1590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 81]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
72.  Kondratyev M, Kreso A, Hallett RM, Girgis-Gabardo A, Barcelon ME, Ilieva D, Ware C, Majumder PK, Hassell JA. Gamma-secretase inhibitors target tumor-initiating cells in a mouse model of ERBB2 breast cancer. Oncogene. 2012;31:93-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 83]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
73.  McGowan PM, Simedrea C, Ribot EJ, Foster PJ, Palmieri D, Steeg PS, Allan AL, Chambers AF. Notch1 inhibition alters the CD44hi/CD24lo population and reduces the formation of brain metastases from breast cancer. Mol Cancer Res. 2011;9:834-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 110]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
74.  Yao J, An Y, Wie JS, Ji ZL, Lu ZP, Wu JL, Jiang KR, Chen P, Xu ZK, Miao Y. Cyclopamine reverts acquired chemoresistance and down-regulates cancer stem cell markers in pancreatic cancer cell lines. Swiss Med Wkly. 2011;141:w13208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
75.  Song Z, Yue W, Wei B, Wang N, Li T, Guan L, Shi S, Zeng Q, Pei X, Chen L. Sonic hedgehog pathway is essential for maintenance of cancer stem-like cells in human gastric cancer. PLoS One. 2011;6:e17687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 129]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
76.  Feldmann G, Fendrich V, McGovern K, Bedja D, Bisht S, Alvarez H, Koorstra JB, Habbe N, Karikari C, Mullendore M. An orally bioavailable small-molecule inhibitor of Hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer. Mol Cancer Ther. 2008;7:2725-2735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 217]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
77.  He B, You L, Uematsu K, Xu Z, Lee AY, Matsangou M, McCormick F, Jablons DM. A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells. Neoplasia. 2004;6:7-14.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Gupta PB, Onder TT, Jiang G, Tao K, Kuperwasser C, Weinberg RA, Lander ES. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 2009;138:645-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1786]  [Cited by in F6Publishing: 1832]  [Article Influence: 122.1]  [Reference Citation Analysis (0)]
79.  Zhi QM, Chen XH, Ji J, Zhang JN, Li JF, Cai Q, Liu BY, Gu QL, Zhu ZG, Yu YY. Salinomycin can effectively kill ALDH(high) stem-like cells on gastric cancer. Biomed Pharmacother. 2011;65:509-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
80.  A Study of Oral LGK974 in Patients With Malignancies Dependent on Wnt Ligands. Cited: 2014-06-27.  Available from: http: //clinicaltrials.gov/ct2/results?term=LGK974&Search=Search.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Clinical trials with Tariquidar (XR9576). Cited: 2014-06-27.  Available from: http: //clinicaltrials.gov/ct2/results?term=tariquidar&Search=Search.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Clinical trials with MS 209. Cited: 2014-06-27.  Available from: http: //clinicaltrials.gov/ct2/results?term=MS-209&Search=Search.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Minderman H, O’Loughlin KL, Pendyala L, Baer MR. VX-710 (biricodar) increases drug retention and enhances chemosensitivity in resistant cells overexpressing P-glycoprotein, multidrug resistance protein, and breast cancer resistance protein. Clin Cancer Res. 2004;10:1826-1834.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  von Furstenberg RJ, Gulati AS, Baxi A, Doherty JM, Stappenbeck TS, Gracz AD, Magness ST, Henning SJ. Sorting mouse jejunal epithelial cells with CD24 yields a population with characteristics of intestinal stem cells. Am J Physiol Gastrointest Liver Physiol. 2011;300:G409-G417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 84]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
85.  Montgomery RK, Carlone DL, Richmond CA, Farilla L, Kranendonk ME, Henderson DE, Baffour-Awuah NY, Ambruzs DM, Fogli LK, Algra S. Mouse telomerase reverse transcriptase (mTert) expression marks slowly cycling intestinal stem cells. Proc Natl Acad Sci USA. 2011;108:179-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 397]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
86.  van der Flier LG, Haegebarth A, Stange DE, van de Wetering M, Clevers H. OLFM4 is a robust marker for stem cells in human intestine and marks a subset of colorectal cancer cells. Gastroenterology. 2009;137:15-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 382]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
87.  van der Flier LG, van Gijn ME, Hatzis P, Kujala P, Haegebarth A, Stange DE, Begthel H, van den Born M, Guryev V, Oving I. Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell. 2009;136:903-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 536]  [Cited by in F6Publishing: 535]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
88.  Formeister EJ, Sionas AL, Lorance DK, Barkley CL, Lee GH, Magness ST. Distinct SOX9 levels differentially mark stem/progenitor populations and enteroendocrine cells of the small intestine epithelium. Am J Physiol Gastrointest Liver Physiol. 2009;296:G1108-G1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 175]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
89.  Okano H, Kawahara H, Toriya M, Nakao K, Shibata S, Imai T. Function of RNA-binding protein Musashi-1 in stem cells. Exp Cell Res. 2005;306:349-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 253]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
90.  May R, Riehl TE, Hunt C, Sureban SM, Anant S, Houchen CW. Identification of a novel putative gastrointestinal stem cell and adenoma stem cell marker, doublecortin and CaM kinase-like-1, following radiation injury and in adenomatous polyposis coli/multiple intestinal neoplasia mice. Stem Cells. 2008;26:630-637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 236]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
91.  Alison MR, Vig P, Russo F, Bigger BW, Amofah E, Themis M, Forbes S. Hepatic stem cells: from inside and outside the liver? Cell Prolif. 2004;37:1-21.  [PubMed]  [DOI]  [Cited in This Article: ]