Editorial Open Access
Copyright ©2009 Baishideng. All rights reserved
World J Stem Cells. Dec 31, 2009; 1(1): 8-10
Published online Dec 31, 2009. doi: 10.4252/wjsc.v1.i1.8
Tumor initiating cells in pancreatic cancer: A critical view
Bo Kong, Christoph W Michalski, Jörg Kleeff, Department of Surgery, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
Author contributions: Kong B, Michalski CW, Kleeff J performed the literature review, analyzed the data, and wrote the paper.
Correspondence to: Jörg Kleeff, MD, Department of Surgery, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany. kleeff@chir.med.tu-muenchen.de
Telephone: +49-89-41405098 Fax: +49-89-4904870
Received: August 26, 2009
Revised: November 11, 2009
Accepted: November 18, 2009
Published online: December 31, 2009

Abstract

Emerging evidence points to the existence of pancreatic cancer stem cells (CSC) as the culprit in the initiation, maintenance, metastasis, and treatment resistance of pancreatic cancer. The existence of such a cell population would have an important impact on the design of novel therapies against this devastating disease. However, no in vivo validation or rebuttal of the pancreatic CSC hypothesis exists. Major backlashes in the discussion on CSC are firstly, the confusion between the terms CSC and cell of origin of pancreatic ductal adenocarcinoma (PDAC), secondly the ambiguity of the cell of origin itself and thirdly, the fact that the CSC hypothesis is based on cell sorting and xenografting experiments; the latter of which often precludes solid conclusions because of the lack of a natural microenvironment and differences in drug delivery. Nonetheless, recent studies in other cancers partially support the CSC hypothesis by demonstrating a link between epithelial-to-mesenchymal transdifferentiation/transition (EMT) and CSC properties. Such a link is again open to dispute as EMT is a reversible process which is highly dependent on major oncogenic pathways in PDAC [e.g. K-Ras, transforming growth factor-β (TGF-β)] rather than on presumed cancer stem cell pathways. Hence, the available evidence does not robustly support the CSC concept in PDAC and a thorough validation of this hypothesis in well-defined genetically engineered mouse models of pancreatic cancer is required.

Key Words: Pancreatic cancer, Cancer stem cell, Tumor initiating cells, Mouse models



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers of the gastrointestinal tract with less than an overall 5-year survival rate of 5%. It is well known that human tumors including PDAC display significant heterogeneity in their respective cell populations. Based on normal hierarchical tissue architecture, the cancer stem cell hypothesis (CSCs) has been developed which suggests that only a specific subset of cancer cells in each tumor is responsible for initiation, maintenance and metastasis[1]. Specifically, the concept implies that a small number of stem or tissue-specific progenitor cancer cells give rise to the terminally differentiated or more committed progeny constituting the bulk mass of cancer, while at the same time these cells also maintain a process of self-renewal[2]. Therefore, following the first characterization of CSCs in acute myeloid leukemia (AML), the identification of pancreatic cancer stem cells was also reported[3,4]. However, a validation of the hypothesis, especially in the context of pancreatic cancer, does not exist. Confusion also derives from imprecise use of the terms “cancer stem cells” and “cell of origin” in PDAC which implies that cancer cells derive from normal tissue stem cells. This is of particular importance in the development of PDAC because the cell or the cellular compartment of origin (i.e. ductal, acinar, centroacinar, endocrine, stem cells) is a subject of controversy. Genetically engineered mouse models of pancreatic ductal adenocarcinoma have made use either of mainly embryonically active, pancreas-specific promoters such as pdx1 or p48 (with specificity towards the exocrine pancreatic cellular compartment) or of inducible constructs driven by, for example, elastase expression to allow for Cre-recombination. Because neither a ductal- nor a centroacinar-specific promoter has been available so far, the cell of origin of PDAC remains obscure. However, initial data from genetically engineered mouse models have suggested that centroacinar cells which are considered to be pancreatic progenitor cells may be the cell of origin of PDAC. In a seminal paper, it has been shown that de-regulation of key pathways (e.g. PI3-K) in centroacinar cells might have contributed to the initiation of mouse PDAC[5,6]. Recently, some reports have argued that adult acinar cells can actually be transformed into pancreatic intraepithelial neoplasia and also into invasive adenocarcinoma , in the presence or in the absence of (chronic) inflammation[7-9]. These papers demonstrate that at least a subset of adult, differentiated cells is readily transformable into (pre-) malignant cells, suggesting that, if existing, a “pancreatic cancer stem cell” is rather derived from a differentiated compartment than from undifferentiated pancreas stem cells.

PANCREATIC CANCER STEM CELLS

So far, the CSC theory in pancreatic cancer largely relies on studies from FACS cell sorting according to the expression of specific “stem-cell” markers (CD133 or CD44/CD24/ESA) followed by xenografting of these cells into immune-compromised mice. Using such a system, a recent study reported that the combined blockade of so-called pancreatic CSC self-renewal pathways and standard chemotherapy eliminated the presumed pancreatic CSCs and resulted in prolonged survival of the transplanted mice[10]. Methodologically, this may not be an appropriate system for testing the CSC theory mainly because xenotransplantation itself has a number of disadvantages. For example, it does not provide the kind of real microenvironment which is usually required for the growth of pancreatic cancer cells[11-14]. Furthermore, drug delivery in xenograft PDAC models has been shown to be completely different than in genetically engineered mouse models[15], underscoring the difficulties in interpreting these results. Thus, the scarcity of so-called “tumor initiating cells” (i.e. CD133+ or CD44+CD24+ESA+) in human PDAC might in fact reflect the scarcity of human tumor cells that can readily adapt to growth in a foreign (mouse) milieu. The non-transplantable human PDAC cells may simply lack critical features for obtaining stromal support in the foreign microenvironment, such as responsiveness to mouse cytokines or chemokines that attract the cells to a nurturing niche rather than “stem-cell” properties[16,17]. Indeed, results from mouse leukemia and lymphoma challenge the general applicability of the cancer stem cell hypothesis to solid tumors because a substantial proportion of cancer cells can seed tumors in syngeneic animals and no functionally distinct subpopulation is evident[18,19]. It has recently also been shown that randomly selected single cells derived from mouse lung or breast cancer cell lines were able to produce tumors after allografting into histocompatible mice[20]. Furthermore, CD133 which is employed as a marker for (pancreatic) CSC is also expressed by endothelial cell precursors which were shown to be capable of enhancing growth of transplanted human cancer cells[21] Thus, using CD133 as a marker to isolate pancreatic cancer stem cells always carries the risk of enriching such a cell population.

EPITHELIAL-TO-MESENCHYMAL TRANSDIFFERENTIATION IN PANCREATIC CANCER

Recent evidence suggests that epithelial-to-mesenchymal transdifferentiation/transition (EMT) in PDAC marks an aggressive and, due to the expression of markers of CSC, a more “cancer stem cell-like” phenotype[22-24]. Accordingly, it has also been shown that highly metastatic pancreatic CSCs loose expression of the epithelial cell marker cytokeratin[3] illustrating a potential link between EMT and the cancer stem cell hypothesis. However, such a link may rather be a side-effect than a true effect since EMT is a reversible process that can be induced by various stimuli [e.g. transforming growth factor (TGF-β)] in the tumor microenvironment[25]. It is likely that in the stepwise malignant transformation process, pancreatic cancer cells have gained an ability to adjust their differentiation status to given environmental influences. This hypothesis seems to be supported by a recent report showing that pancreatic cancers can be divided into K-ras-dependent and -independent tumors. A comparison of these two classes of cancer cells revealed a gene expression signature in K-ras-dependent cells that was associated with a well-differentiated epithelial phenotype[26]. However, no changes in CSC marker expression has been reported after induction of EMT in such K-ras-dependent pancreatic cancer cells. Thus, the CSC “population” may also be considered as a transient state of the parental cancer cells which again would argue against the central concept of the cancer stem cell hypothesis.

CONCLUSION

Though the determination of the validity of the pancreatic CSC hypothesis would have an important impact on the design of novel therapies, the available evidence does not robustly support the CSC concept in PDAC. Therefore, we suggest analyzing the concept in well-defined genetically engineered mouse models of pancreatic cancer with the sole aim of eradicating the hypothesized minor population of CSC. Such experiments would determine whether ablation of this presumed population of tumor-initiating cells has an effect on the development and potentially also the progression of the tumor.

Footnotes

Peer reviewer: Ursula Margarethe Gehling, MD, Paul-Ehrlich-Institute, Paul-Ehrlich-Strasse 51 - 59, 63225 Langen, Germany

S- Editor Li LF L- Editor Hughes D E- Editor Lin YP

References
1.  Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645-648.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Bednar F, Simeone DM. Pancreatic cancer stem cells and relevance to cancer treatments. J Cell Biochem. 2009;107:40-45.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, Bruns CJ, Heeschen C. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell. 2007;1:313-323.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  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: ]
5.  Esposito I, Seiler C, Bergmann F, Kleeff J, Friess H, Schirmacher P. Hypothetical progression model of pancreatic cancer with origin in the centroacinar-acinar compartment. Pancreas. 2007;35:212-217.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Stanger BZ, Stiles B, Lauwers GY, Bardeesy N, Mendoza M, Wang Y, Greenwood A, Cheng KH, McLaughlin M, Brown D. Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. Cancer Cell. 2005;8:185-195.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291-302.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  De La O JP, Emerson LL, Goodman JL, Froebe SC, Illum BE, Curtis AB, Murtaugh LC. Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc Natl Acad Sci USA. 2008;105:18907-18912.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Habbe N, Shi G, Meguid RA, Fendrich V, Esni F, Chen H, Feldmann G, Stoffers DA, Konieczny SF, Leach SD. Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice. Proc Natl Acad Sci USA. 2008;105:18913-18918.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Mueller MT, Hermann PC, Witthauer J, Rubio-Viqueira B, Leicht SF, Huber S, Ellwart JW, Mustafa M, Bartenstein P, D'Haese JG. Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology. 2009;137:1102-1113.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57-70.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Erkan M, Kleeff J, Gorbachevski A, Reiser C, Mitkus T, Esposito I, Giese T, Büchler MW, Giese NA, Friess H. Periostin creates a tumor-supportive microenvironment in the pancreas by sustaining fibrogenic stellate cell activity. Gastroenterology. 2007;132:1447-1464.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Kleeff J, Beckhove P, Esposito I, Herzig S, Huber PE, Löhr JM, Friess H. Pancreatic cancer microenvironment. Int J Cancer. 2007;121:699-705.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Arai KI, Lee F, Miyajima A, Miyatake S, Arai N, Yokota T. Cytokines: coordinators of immune and inflammatory responses. Annu Rev Biochem. 1990;59:783-836.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457-1461.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Adams JM, Strasser A. Is tumor growth sustained by rare cancer stem cells or dominant clones? Cancer Res. 2008;68:4018-4021.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Bhagwandin VJ, Shay JW. Pancreatic cancer stem cells: fact or fiction? Biochim Biophys Acta. 2009;1792:248-259.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Kelly PN, Dakic A, Adams JM, Nutt SL, Strasser A. Tumor growth need not be driven by rare cancer stem cells. Science. 2007;317:337.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Somervaille TC, Cleary ML. Identification and characterization of leukemia stem cells in murine MLL-AF9 acute myeloid leukemia. Cancer Cell. 2006;10:257-268.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Yoo MH, Hatfield DL. The cancer stem cell theory: is it correct? Mol Cells. 2008;26:514-516.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Bruno S, Bussolati B, Grange C, Collino F, Graziano ME, Ferrando U, Camussi G. CD133+ renal progenitor cells contribute to tumor angiogenesis. Am J Pathol. 2006;169:2223-2235.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S, Abbruzzese JL, Gallick GE, Sarkar FH. Acquisition of epithelial-mesenchymal transition phenotype of gemcitabine-resistant pancreatic cancer cells is linked with activation of the notch signaling pathway. Cancer Res. 2009;69:2400-2407.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704-715.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Shah AN, Summy JM, Zhang J, Park SI, Parikh NU, Gallick GE. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 2007;14:3629-3637.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Jungert K, Buck A, von Wichert G, Adler G, König A, Buchholz M, Gress TM, Ellenrieder V. Sp1 is required for transforming growth factor-beta-induced mesenchymal transition and migration in pancreatic cancer cells. Cancer Res. 2007;67:1563-1570.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Singh A, Greninger P, Rhodes D, Koopman L, Violette S, Bardeesy N, Settleman J. A gene expression signature associated with "K-Ras addiction" reveals regulators of EMT and tumor cell survival. Cancer Cell. 2009;15:489-500.  [PubMed]  [DOI]  [Cited in This Article: ]