Sei sulla pagina 1di 7

Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.

CAN-10-3220

Cancer
Review Research

Cancer Stem Cell Niche: The Place to Be


Tijana Borovski, Felipe De Sousa E Melo, Louis Vermeulen, and Jan Paul Medema

Abstract
Tumors are being increasingly perceived as abnormal organs that, in many respects, recapitulate the
outgrowth and differentiation patterns of normal tissues. In line with this idea is the observation that only a
small fraction of tumor cells is capable of initiating a new tumor. Because of the features that these cells share with
somatic stem cells, they have been termed cancer stem cells (CSC). Normal stem cells reside in a "stem cell niche"
that maintains them in a stem-like state. Recent data suggest that CSCs also rely on a similar niche, dubbed the
"CSC niche," which controls their self-renewal and differentiation. Moreover, CSCs can be generated by the
microenvironment through induction of CSC features in more differentiated tumor cells. In addition to a role in
CSC maintenance, the microenvironment is hypothesized to be involved in metastasis by induction of the
epithelial-mesenchymal transition, leading to dissemination and invasion of tumor cells. The localization of
secondary tumors also seems to be orchestrated by the microenvironment, which is suggested to form a
premetastatic niche. Thus, the microenvironment seems to be of crucial importance for primary tumor growth as
well as metastasis formation. Combined with its role in the protection of CSCs against genotoxic insults, these
data strongly put forward the niche as an important target for novel therapies. Cancer Res; 71(3); 634–9. 2011 AACR.

Introduction cells and the nonmalignant cells that make up the tumor
environment. Like normal stem cells, CSCs seem to depend on
It is becoming increasingly clear that tumors are hierarchi- a similar, permissive environment, the CSC niche, to retain
cally organized heterogeneous populations of cells with the their exclusive abilities to self-renew and give rise to more
cancer stem cell (CSC) compartment on top. This fraction of differentiated progenitor cells, while staying in an undiffer-
tumor cells shares many similarities with normal stem cells, entiated state themselves (5). Moreover, the CSC niche also
such as self-renewing capacity and multilineage differentia- has a protective role. By sheltering CSCs from diverse geno-
tion properties (1). In addition, CSCs are highly tumorigenic toxic insults, the niche contributes to their enhanced therapy
and can generate a serially transplantable phenocopy of the resistance (6, 7). Here, we discuss the concept of the stem cell
primary human malignancy in immunocompromised mice niche in a tumor setting, with special emphasis on brain and
(1). From a clinical point of view, the main concern with CSCs colon cancer as examples of malignancies in which CSCs seem
is their resistance to conventional treatments, a feature sug- to rely on a specialized microenvironment. Furthermore, we
gested to be the underlying cause of tumor recurrence (2, 3). review the role of the tumor microenvironment in the pro-
Thus, it is necessary to define the factors that sustain CSCs in gression of primary tumors, focusing on dedifferentiation of
order to develop more efficient therapeutics. Normal stem non-CSCs and epithelial-mesenchymal transition (EMT)
cells reside in the distinct environment called the "stem cell induction by the niche and, finally, the involvement of a
niche." The niche regulates stemness, proliferation, and apop- premetastatic niche in the formation of metastasis (Fig. 1).
tosis resistance of stem cells. It has a complex architecture and
is composed of diverse stromal cells, such as mesenchymal The Perivascular Niche in Glioblastoma
and immune cells, a vascular network, soluble factors, and Multiforme
extracellular matrix components. Analogously, tumorigenicity
not only involves the biology of tumor cells themselves but In the adult mammalian brain, neural stem cells reside in
also results from a rather complex interplay between tumor the hippocampus and subventricular zone, close to the blood
vessels. During embryogenesis and early brain development,
ventricular neuroectoderm secretes high levels of vascular
Authors' Affiliations: Laboratory for Experimental Oncology and Radio- endothelial growth factor (VEGF), which attracts and stimu-
biology (LEXOR), Center for Experimental Molecular Medicine (CEMM), lates vessel growth in this region of the brain (8). Thus,
Academic Medical Center (AMC), University of Amsterdam, Amsterdam,
the Netherlands vascular endothelial cells and neural stem cells come together
Corresponding Author: Jan Paul Medema, AMC, Meibergdreef 9, Room during early development and stay in close proximity through-
G2–131, Amsterdam, 1105AZ, the Netherlands. Phone: 31–20-5667777; out life. Protein ligands found within the neural stem cell
Fax: 31–20-6977192; E-mail: J.P.Medema@amc.uva.nl niche, such as pigment epithelium–derived factor and stem
doi: 10.1158/0008-5472.CAN-10-3220 cell factor, have been implicated in both stem cell self-renewal
2011 American Association for Cancer Research. and regulation of angiogenesis, also suggesting that these two

634 Cancer Res; 71(3) February 1, 2011

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

The Cancer Stem Cell Niche

Figure 1. The CSC niche in tumor growth and metastasis. The CSC niche is composed of blood vessels (red), stromal cells such as myofibroblasts
(orange), and extracellular matrix components. Tumors are organized in such a way that CSCs (purple) reside close to their niche. In addition to maintaining
CSCs in a stemlike state, (1), the niche has the ability to dedifferentiate nontumorigenic cells (blue) into tumorigenic CSCs (purple) and, (2), to induce the
EMT, leading to dissemination of tumor cells from the primary tumor and, (3), seeding at the metastatic place. Furthermore, tumor cell engraftment in
different organs is suggested to be facilitated by the formation of a premetastatic niche that potentially enables the initiation and outgrowth of secondary
tumors.

processes are tightly linked (9–12). More direct evidence for vessels and, furthermore, that the vascular endothelial cells
the role of endothelial cells in neural stem cell biology comes are able to maintain patient-derived brain tumor cells in a
from the observation that these cells regulate asymmetrical stemlike state and promote their tumorigenicity when coin-
division of the stem cells in the subventricular zone (13). jected in immunocompromised mice (5). A soluble factor
Furthermore, they maintain neural stem cell self-renewal, in mediating this interaction and promoting CSC self-renewal
part via Notch signaling, and simultaneously inhibit stem cell seems to be nitric oxide produced by endothelial cells, which
differentiation (4). activates the Notch pathway in glioma CSCs (14). However, the
Glioblastoma multiforme (GBM) is the most aggressive type interaction between CSCs and tumor vasculature is likely a
of primary brain tumor in humans, with high morbidity and more complex and bidirectional process. Brain tumor CSCs
median survival of less then a year. Recently, a CSC fraction are able to promote recruitment and formation of blood
was identified in these tumors. These glioma-initiating cells vessels by secreting VEGF (15). Furthermore, inhibition of
seem to have a higher DNA repair rate and resistance to angiogenesis and depletion of blood vessels by the VEGF-
treatment than more differentiated glioma cells, making them neutralizing antibody bevacizumab reduced the CSC pool and,
the main suspects for tumor regrowth after therapy (2). GBM subsequently, inhibited tumor growth (5). Moreover, tumor
is a highly vascularized tumor, which led to the speculation cells protect their niche and, vice versa, the vascular micro-
that GBM CSCs might depend on a similar niche as neural environment contributes to enhanced therapy resistance of
stem cells. Indeed, recent publications have shown the validity GBM CSCs. Accordingly, glioma cells induce upregulation
of this concept. Calabrese and colleagues showed the exis- of different survival genes in endothelial cells, protecting
tence of a close relationship between brain CSCs and blood them from hypoxia or irradiation-induced apoptosis (16, 17).

www.aacrjournals.org Cancer Res; 71(3) February 1, 2011 635

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

Borovski et al.

Reciprocally, inhibition of Notch signaling in an explant system expression leads to complete loss of crypts in adult mice
of surgical GBM specimens leads to detachment of CSCs from (23). BMP signaling has opposite effects and drives the cells
their vascular niche and increased efficacy of radiotherapy on toward differentiation (24). A fine-tuned balance between
CSCs (6). Similarly, application of antiangiogenic therapy to these and other signaling pathways maintains the intestinal
gliomas and eradication of tumor vasculature results in a homeostasis and this is partially directed by the intestinal
higher susceptibility of CSCs to cytotoxic agents (7). Although stem cell niche. Myofibroblasts that line the crypt produce
these data sound promising, they should be viewed with Wnt ligands, together with BMP antagonists such as gremlin
caution as the long-term effect of antiangiogenic therapy on 1/2, which, in combination, are involved in preservation of the
tumor growth and the final outcome of the treatment is still not stem cell pool (25, 26).
fully known. A recent publication even suggested that anti- A similar model has been suggested to delineate the colon
VEGF treatment eventually leads to increased invasiveness and CSC interaction with their microenvironment. Studies of
metastasis of the tumor (18). One of the undesired conse- heritable juvenile polyposis syndrome highlighted the impor-
quences of anti-VEGF treatment could be an increase in tance of tumor stroma for the development of colorectal
hypoxic areas, which are described to be refractory to therapy. cancer. One of the features of the gastrointestinal polyps seen
Being located far from blood vessels, hypoxic tumor cells are in Apc-Smad4 mutant heterozygous mice is increased prolif-
usually exposed to relatively low concentrations of chemother- eration of stromal cells. It is also one of the characteristics of
apeutics. Furthermore, cytotoxic drugs mainly target prolifer- juvenile polyps seen in humans and predisposes for develop-
ating cells and, therefore, do not harm hypoxic tumor cells that ment of carcinomas that arise from epithelial cells. In addi-
are usually quiescent due to a lack of oxygen and nutrients (19). tion, a connection between inflammatory processes and colon
In addition, hypoxia also creates problems for radiotherapy, as cancer is well established. For example, it has been described
it reduces the formation of oxygen free radicals that, as a in models of colitis-associated cancer, as well as in other forms
byproduct of radiotherapy, would normally induce DNA of colorectal cancer, that interleukin 6 secreted mainly by the
damage in tumor cells (20). Hypoxia also has more direct infiltrating immune cells stimulates proliferation of tumor-
cellular effects, as it has been linked to regulating cell survival, initiating intestinal cells (27). Analogously to the normal
enhanced motility and invasiveness of tumor cells, and tumor intestinal stem cell niche, myofibroblasts and mesenchymal
angiogenesis. Moreover, hypoxic regions are related to the stem cells are shown to be components of the colon cancer
areas of pseudopallisading necrosis, another hallmark of stroma. Mesenchymal stem cells have the ability to enhance
GBM, and have been proposed to also form a GBM CSC niche, growth and metastasis of colon cancer and have been, further-
in addition to tumor vasculature (21). GBM CSCs are more, proposed to give rise to fibroblasts that further promote
frequently found to be located at the edges of necrotic regions. tumorigenesis (28, 29). Mutations in the APC gene, an inhibitor
Indeed, similar to normal neural stem cells, it has also been of Wnt signaling, are early events in the transition of healthy
shown that hypoxia regulates GBM CSC maintenance (21, 22). colon mucosa toward colon carcinoma, resulting in stabiliza-
The effects of hypoxia are mainly mediated by hypoxia-indu- tion of b-catenin and subsequent translocation to the nucleus.
cible factors (HIF), of which HIF2a is of particular importance In the nucleus, b-catenin binds to T-cell factor/lymphoid
for the GBM CSC pool, increasing their self-renewal and enhancer factor (TCF/LEF) family members and acts as a
tumorigenic capacity (21). Not surprisingly, HIF2a expression transcriptional regulator of many genes that control prolif-
correlates with the poor survival of GBM patients. Combined, eration and differentiation. However, despite the same genetic
these data clearly put forward the role of brain tumor background, cells within the tumors display differential Wnt
vasculature and hypoxia in the maintenance of CSCs, as well activity, judging by the localization of b-catenin, which is
as their therapy resistance; however, further investigations are referred to as the so-called b-catenin paradox (30, 31). It
needed to successfully apply this knowledge in treating GBM indicates that, besides APC mutations, additional regulatory
patients. mechanisms of Wnt activity are at play in these tumors. One
possible explanation could be additional KRAS mutations.
The Stem Cell Niche in Colorectal Cancer According to Phelps and colleagues, APC mutations primarily
play a role in stabilizing the levels of b-catenin in the cyto-
Another example of an extensively studied stem cell niche plasm, whereas additional mutations in KRAS are necessary
exists in the intestine. The intestinal crypt is the functional for translocation of b-catenin to the nucleus (32). In addition
unit of the intestinal tract, including colon and small intestine. to these cell-intrinsic events, the Wnt pathway also seems to
Stem cells reside in the bottom region of the crypt, within a be regulated by the tumor microenvironment. Accordingly,
stem cell niche composed of epithelial cells and mesenchymal cells harboring nuclear b-catenin are mainly clustered at the
cells of the myofibroblast lineage that line the crypt. Moving invasive front of the tumor in regions highly populated with
upwards, progenitor cells start to differentiate and once they myofibroblasts (30, 31). How does this finding relate to colon
reach the top, they are shed into the lumen of intestine and CSCs? Tumor-associated myofibroblasts (TAF) are a major
die. Current data show that the Wnt signaling cascade is a cellular component of colon cancer stroma. They have higher
prominent force controlling cell proliferation, differentiation, proliferation rates than normal fibroblasts and are the primary
and apoptosis along the crypt–villus axis and in maintaining source of type I collagen, shown to promote a stem cell–like
stem cell fate (23). The absence of Wnt signaling activity in phenotype in colorectal carcinoma cell lines (33). The impor-
Tcf4/ mice or its inhibition by transgenic Dickkopf-1 tance of TAFs in tumorigenesis is supported by data showing

636 Cancer Res; 71(3) February 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

The Cancer Stem Cell Niche

that depletion of TAFs through CD8þ T-cell–mediated killing stem cell factors, such as OCT4, NANOG, and c-MYC (38).
significantly reduced tumor growth and metastasis in, among Furthermore, expression of HIF2a in the non–stem cell popu-
others, colon cancer (34). In addition, efficacy of chemother- lation enhanced their tumorigenic potential. It should be
apy was improved, implying that TAFs are also involved in noted, though, that, in addition to regulating cell plasticity,
tumor chemoresistance. Recently, we reported that high Wnt hypoxia, and other EMT-inducing factors from the microen-
activity levels mark the colon CSC population and are orche- vironment, such as TGF-b, are also known to stimulate
strated by myofibroblasts residing in the tumor microenvir- proliferation and expansion of the preexisting CSC pool,
onment (35). More precisely, we found that hepatocyte growth thereby further increasing the chances for metastatic spread
factor (HGF) produced by myofibroblasts is capable of enhan- (37, 39, 40). One of the mechanisms of hypoxia-induced
cing Wnt signaling activity in colon CSCs, suggesting a strong invasiveness is the activation of the Wnt signaling pathway
link between the microenvironment and CSC features in via inhibition of glycogen synthase kinase 3b, which leads to
colorectal cancer as well. the induction of the key EMT-inducing transcription factor
Snail (41). Expression of SNAIL protein was detected at the
Cancer Stem Cells, Dedifferentiation, and the tumor–stroma interface in diverse human cancers including
Epithelial-Mesenchymal Transition colon cancer (42). Correspondingly, it was mentioned pre-
viously that colorectal cancer cells with nuclear b-catenin, a
Intriguingly, the effects of the microenvironment on CSCs marker of colon CSCs on one hand and an inducer of EMT on
are beyond just preservation and protection of this compart- the other, mostly reside at the host–tumor interface (30).
ment. We observed that HGF-producing myofibroblasts Intriguingly, HGF, which we have described as being able
were able to dedifferentiate nontumorigenic tumor cells into to induce CSC properties in differentiated colon cancer cells,
more immature cells by reactivating the Wnt pathway. These was used to induce cell scattering of MDCK cells in the initial
dedifferentiated cancer cells displayed all characteristics of studies on EMT (43). The ability of reacquiring stem cell
CSCs, including expression of stem cell–associated genes, features in the more differentiated cells by HGF-producing
such as LGR5, and high tumorigenic potential (35). Our myofibroblasts puts these cells at the crucial position in the
experiments imply that the tumor microenvironment, more EMT-CSC framework.
specifically the CSC niche, is capable of inducing a CSC
phenotype in differentiated tumor cells. The Premetastatic Niche
What would be the consequences of an ongoing process of
dedifferentiation in malignancies? Cancer cells possess a We have described how TAFs are involved in maintenance
certain level of plasticity that allows them to change their of CSCs and the induction of CSC features in more differ-
phenotype and acquire different functions and properties entiated tumor cells, potentially via mechanisms related to
under the influence of the environment. Processes that reflect EMT. Moreover, we have put forward how this model provides
their plasticity are the EMT and its reverse, the mesenchymal- an elegant explanation for the intimate connection between
epithelial transition, highly conserved programs that are CSC features, the EMT, and tumor cell invasion. However, the
involved in embryonic development but also in carcinogenesis microenvironment might also be implicated in the final steps
(36). EMT is one of the crucial, early steps in the invasion– of the metastatic cascade. It is known that metastases selec-
metastasis cascade and has been associated with poor clinical tively occur in certain organs such as lungs, liver, brain, and
outcome of patients in many types of tumors. Epithelial tumor bones. This observation led to the so-called seed and soil
cells that undergo EMT lose cell–cell adhesion properties and hypothesis. According to this hypothesis, the local microen-
polarity and acquire a more mesenchymal-like phenotype, vironment of these organs seems to be more receptive to
including motility, invasiveness, and increased resistance to disseminated tumor cells from particular malignancies than
apoptosis. Importantly, tumor cells undergoing EMT acquire other organs. Thus, occurrence of metastasis does not happen
CSC-like features as can be concluded from experiments in randomly, but disseminated tumor cells need to meet a
which induction of EMT in immortalized human mammary hospitable microenvironment in order to initiate a secondary
epithelial cells led to the expression of CSC markers, increased tumor. Furthermore, in recent years, evidence suggested that
self-renewal capacity, and enhanced tumor formation (37). the primary tumor itself is actively involved in adapting these
EMT, thus, provides disseminated tumor cells with self-renew- so-called premetastatic niches for tumor cells to come, by
ing properties and increased proliferative capacity, enhancing secreting systemic factors and directing bone marrow–
their chances to seed at a distant site and grow metastases. derived cells and macrophages to certain tissues, thereby
Considering the fact that the EMT promotes the generation of priming certain tissues for tumor cell engraftment (44, 45).
a CSC phenotype, it is crucial to know how this process is Accordingly, VEGFR1-positive bone marrow–derived hema-
controlled and regulated. A variety of signals that can induce topoietic progenitor cells (HPC) were shown to localize to
the EMT, such as hypoxia, are being received from the tumor premetastatic sites and form clusters before the arrival of
microenvironment. Hypoxia was shown to regulate the plas- tumor cells (44). Eradication of these cells from the bone
ticity of GBM cells as well. Heddleston and colleagues found marrow prevents the formation of premetastatic clusters and,
hypoxia to promote the self-renewal capability and stem cell subsequently, tumor metastasis. In addition to homing of
phenotype in the non–stem cell population, increasing their HPCs, preexisting fibroblasts are noted to increase fibronectin
neurosphere-forming capacity and upregulating important deposition on these sites, which most likely binds to VLA4, a

www.aacrjournals.org Cancer Res; 71(3) February 1, 2011 637

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

Borovski et al.

fibronectin receptor expressed on HPCs, and facilitates accu- ing metastasis formation, clearly puts the CSC niche and,
mulation of these cells. Furthermore, activated fibroblasts especially, the mediators of this interaction in the spotlight as
were shown to induce remodeling of stroma required for liver future therapeutic targets. Emerging therapies are already
metastasis in a murine melanoma model (46). Thus, in addi- targeting this strategy. For example, we previously highlighted
tion to their contribution to the CSC niche at the primary the importance of HGF production by the microenvironment
tumor site, fibroblasts are suggested to have a critical role in in colorectal cancer. HGF acts via the tyrosine kinase receptor
premetastasis niche formation as well. MET on cancer cells and triggers its downstream targets.
Recently, it has been shown that anti-MET antibodies prevent
Conclusions HGF binding to MET and, subsequently, inhibit colon cancer
tumor growth (47). This example is only one of the extensive
In conclusion, the reviewed data point out a central role of studies that explore the modulation of the interaction
the CSC niche in virtually every step of the tumorigenic between cancer cells and niche cells as a therapeutic strategy
cascade (Fig. 1). In primary tumors, the CSC niche is an that could lead to major advances in cancer treatment in the
important regulator of stemness. The importance of this years to come.
interaction is supported by the fact that the loss of a niche
environment mainly leads to the loss of CSCs. The reliance of Disclosure of Potential Conflicts of Interest
CSCs on niche signals seems to be a general phenomenon and
has been shown in a whole variety of different tumors. In No potential conflicts of interest were disclosed.
addition to maintaining the CSC pool and supporting the
growth of primary tumors, the niche plays a role in reverting Acknowledgments
nontumorigenic cells into CSCs by processes related to the
EMT, leading to tumor invasion and dissemination. Finally, The authors would like to thank the members of the group for useful
the putative premetastatic niche supposedly assures success- discussions.
ful homing of cancer cells to distant organs and the devel-
opment of metastasis. Whether premetastatic niches are also Grant Support
capable of installing a CSC phenotype in more differentiated
J.P. Medema is supported by a VICI grant from the Dutch Science Organiza-
cells or whether these particular environments are only cap- tion and through a grant from the Dutch Cancer Society (UvA2009–4416).
able of maintaining the function of metastasized CSCs is still a
matter of speculation. Either way, the supporting role of the Received September 2, 2010; revised November 8, 2010; accepted November
microenvironment in tumor growth and progression, includ- 11, 2010; published OnlineFirst January 25, 2011.

References
1. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. 10. Jin K, Mao XO, Sun YJ, Xie L, Greenberg DA. Stem cell factor
Cancer stem cells–perspectives on current status and future direc- stimulates neurogenesis in vitro and in vivo. J Clin Invest
tions: AACR Workshop on cancer stem cells. Cancer Res 2002;110:311–9.
2006;66:9339–44. 11. Ramirez-Castillejo C, Sanchez-Sanchez F, Andreu-Agullo C, Ferron
2. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. SR, Aroca-Aguilar JD, Sanchez P, et al. Pigment epithelium-derived
Glioma stem cells promote radioresistance by preferential activation factor is a niche signal for neural stem cell renewal. Nat Neurosci
of the DNA damage response. Nature 2006;444:756–60. 2006;9:331–9.
3. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Lovino 12. Dawson DW, Volpert OV, Gillis P, Crawford SE, Xu HJ, Benedict W,
F, et al. Colon cancer stem cells dictate tumor growth and resist cell et al. Pigment epithelium-derived factor: A potent inhibitor of angio-
death by production of interleukin-4. Cell Stem Cell 2007;1:389–402. genesis. Science 1999;285:245–8.
4. Shen Q, Goderie SK, Jin L, Karanth N, Sun Y, Abramova N, et al. 13. Knoblich JA. Mechanisms of asymmetric stem cell division. Cell
Endothelial cells stimulate self-renewal and expand neurogenesis of 2008;132:583–97.
neural stem cells. Science 2004;304:1338–40. 14. Charles N, Ozawa T, Squatrito M, Bleau AM, Brennan CW, Hambard-
5. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, zumyan D, et al. Perivascular nitric oxide activates notch signaling and
et al. A perivascular niche for brain tumor stem cells. Cancer Cell promotes stem-like character in PDGF-induced glioma cells. Cell
2007;11:69–82. Stem Cell 2010;6:141–52.
6. Hovinga KE, Shimizu F, Wang R, Panagiotakos G, Van Der Heijden M, 15. Bao S, Wu Q, Sathornsumetee S, Hao YL, Li ZZ, Hjelmeland AB,
Moayedpardazi H, et al. Inhibition of Notch signaling in glioblastoma et al. Stem cell-like glioma cells promote tumor angiogenesis
targets cancer stem cells via an endothelial cell intermediate. Stem through vascular endothelial growth factor. Cancer Res 2006;66:
Cells 2010;28:1019–29. 7843–8.
7. Folkins C, Man S, Xu P, Shaked Y, Hicklin DJ, Kerbel RS. Anticancer 16. Ezhilarasan R, Mohanam I, Govindarajan K, Mohanam S. Glioma cells
therapies combining antiangiogenic and tumor cell cytotoxic effects suppress hypoxia-induced endothelial cell apoptosis and promote the
reduce the tumor stem-like cell fraction in glioma xenograft tumors. angiogenic process. Int J Oncol 2007;30:701–7.
Cancer Res 2007;67:3560–4. 17. Brown CK, Khodarev NN, Yu JQ, Moo-Young T, Labay E, Darga TE,
8. Breier G, Albrecht U, Sterrer S, Risau W. Expression of vascular et al. Glioblastoma cells block radiation-induced programmed cell
endothelial growth-factor during embryonic angiogenesis and death of endothelial cells. FEBS Lett 2004;565:167–70.
endothelial cell differentiation. Development 1992;114:521–32. 18. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F,
9. Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S, et al. et al. Antiangiogenic therapy elicits malignant progression of tumors
Neuronal and glioma-derived stem cell factor induces angiogenesis to increased local invasion and distant metastasis. Cancer Cell
within the brain. Cancer Cell 2006;9:287–300. 2009;15:220–31.

638 Cancer Res; 71(3) February 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

The Cancer Stem Cell Niche

19. Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev 34. Loeffler M, Kruger JA, Niethammer AG, Reisfeld RA. Targeting tumor-
Cancer 2006;6:583–92. associated fibroblasts improves cancer chemotherapy by increasing
20. Hockel M, Vaupel P. Tumor hypoxia: definitions and current clinical, intratumoral drug uptake. J Clin Invest 2006;116:1955–62.
biologic, and molecular aspects. J Natl Cancer Inst 2001;93:266–76. 35. Vermeulen L, De Sousa E Melo F, Van Der Heijden M, Cameron K, de
21. Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, et al. Hypoxia- Jong JH, Borovski T, et al. Wnt activity defines colon cancer stem cells
inducible factors regulate tumorigenic capacity of glioma stem cells. and is regulated by the microenvironment. Nat Cell Biol 2010;12:468–
Cancer Cell 2009;15:501–13. 76.
22. Clarke L, Van Der Kooy D. Low oxygen enhances primitive and 36. Yang J, Weinberg RA. Epithelial-mesenchymal transition: At the cross-
definitive neural stem cell colony formation by inhibiting distinct cell roads of development and tumor metastasis. Dev Cell 2008;14:818–29.
death pathways. Stem Cells 2009;27:1879–86. 37. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The
23. Clevers H. Wnt/beta-catenin signaling in development and disease. epithelial-mesenchymal transition generates cells with properties of
Cell 2006;127:469–80. stem cells. Cell 2008;133:704–15.
24. He XC, Zhang JW, Tong WG, Tawfik O, Ross J, Scoville DH, et al. 38. Heddleston JM, Li ZZ, McLendon RE, Hjelmeland AB, Rich JN. The
BMP signaling inhibits intestinal stem cell self-renewal through hypoxic microenvironment maintains glioblastoma stem cells and
suppression of Wnt-beta-catenin signaling. Nat Genet 2004;36: promotes reprogramming towards a cancer stem cell phenotype. Cell
1117–21. Cycle 2009;8:3274–84.
25. Kosinski C, Li VSW, Chan ASY, Zhang J, Ho C, Tsui WY, et al. Gene 39. Blazek ER, Foutch JL, Maki G. Daoy medulloblastoma cells that express
expression patterns of human colon tops and basal crypts and BMP CD133 are radioresistant relative to CD133- cells, and the CD133þ
antagonists as intestinal stem cell niche factors. Proc Natl Acad Sci sector is enlarged by hypoxia. Int J Radiat Oncol Biol Phys 2007;67:1–5.
U S A 2007;104:15418–23. 40. Das B, Tsuchida R, Malkin D, Koren G, Baruchel S, Yeger H. Hypoxia
26. Gregorieff A, Pinto D, Begthel H, Destree O, Kielman M, Clevers H. enhances tumor stemness by increasing the invasive and tumorigenic
Expression pattern of Wnt signaling components in the adult intestine. side population fraction. Stem Cells 2008;26:1818–30.
Gastroenterology 2005;129:626–38. 41. Cannito S, Novo E, Compagnone A, Valfre di Bonzo L, Busletta C,
27. Grivennikov S, Karin E, Terzic J, Mucida D, Yu GY, Vallabhapurapu S, Zamara E, et al. Redox mechanisms switch on hypoxia-dependent
et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells epithelial-mesenchymal transition in cancer cells. Carcinogenesis
and development of colitis-associated cancer. Cancer Cell 2009;15: 2008;29:2267–78.
103–13. 42. Franci C, Takkunen M, Dave N, Alameda F, Gomez S, Rodriguez R,
28. Shinagawa K, Kitadai Y, Tanaka M, Sumida T, Kodama M, Higashi Y, et al. Expression of Snail protein in tumor-stroma interface. Oncogene
et al. Mesenchymal stem cells enhance growth and metastasis of 2006;25:5134–44.
colon cancer. Int J Cancer 2010;127:2323–33. 43. Stoker M, Perryman M. An epithelial scatter factor released by embryo
29. Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, fibroblasts. J Cell Sci 1985;77:209–23.
et al. Carcinoma-associated fibroblast-like differentiation of human 44. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C,
mesenchymal stem cells. Cancer Res 2008;68:4331–9. et al. VEGFR1-positive haematopoietic bone marrow progenitors
30. Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz-Schughart LA, initiate the pre-metastatic niche. Nature 2005;438:820–7.
et al. Variable beta-catenin expression in colorectal cancers indicates 45. Hiratsuka S, Watanabe A, Aburatani H, Maru Y. Tumour-mediated
tumor progression driven by the tumor environment. Proc Natl Acad upregulation of chemoattractants and recruitment of myeloid cells
Sci U S A 2001;98:10356–61. predetermines lung metastasis. Nat Cell Biol 2006;8:1369–75.
31. Fodde R, Brabletz T. Wnt/beta-catenin signaling in cancer stemness 46. Olaso E, Santisteban A, Bidaurrazaga J, Gressner AM, Rosenbaum J,
and malignant behavior. Curr Opin Cell Biol 2007;19:150–8. Vidal-Vanaclocha F. Tumor-dependent activation of rodent hepatic
32. Phelps RA, Chidester S, Dehghanizadeh S, Phelps J, Sandoval IT, Rai stellate cells during experimental melanoma metastasis. Hepatology
K, et al. A two-step model for colon adenoma initiation and progres- 1997;26:634–42.
sion caused by APC loss. Cell 2009;137:623–34. 47. van der Horst EH, Chinn L, Wang M, Velilla T, Tran H, Madrona Y, et al.
33. Kirkland SC. Type I collagen inhibits differentiation and promotes a Discovery of fully human anti-MET monoclonal antibodies with anti-
stem cell-like phenotype in human colorectal carcinoma cells. Br J tumor activity against colon cancer tumor models in vivo. Neoplasia
Cancer 2009;101:320–6. 2009;11:355–64.

www.aacrjournals.org Cancer Res; 71(3) February 1, 2011 639

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.
Published OnlineFirst January 25, 2011; DOI: 10.1158/0008-5472.CAN-10-3220

Cancer Stem Cell Niche: The Place to Be


Tijana Borovski, Felipe De Sousa E Melo, Louis Vermeulen, et al.

Cancer Res 2011;71:634-639. Published OnlineFirst January 25, 2011.

Updated version Access the most recent version of this article at:
doi:10.1158/0008-5472.CAN-10-3220

Cited articles This article cites 47 articles, 10 of which you can access for free at:
http://cancerres.aacrjournals.org/content/71/3/634.full#ref-list-1

Citing articles This article has been cited by 11 HighWire-hosted articles. Access the articles at:
http://cancerres.aacrjournals.org/content/71/3/634.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://cancerres.aacrjournals.org/content/71/3/634.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's
(CCC)
Rightslink site.

Downloaded from cancerres.aacrjournals.org on June 8, 2018. © 2011 American Association for Cancer
Research.

Potrebbero piacerti anche