You are on page 1of 10

IJC

International Journal of Cancer

Tumor heterogeneity and cancer stem cell paradigm: Updates


in concept, controversies and clinical relevance
Anup Kumar Singh1*, Rakesh Kumar Arya1*, Shrankhla Maheshwari1,2*, Akhilesh Singh1,
Sanjeev Meena1, Priyanka Pandey3, Olivier Dormond4 and Dipak Datta1,2
1
Biochemistry Division, CSIR-Central Drug Research Institute, Lucknow, India
2
Academy of Scientific and Innovative Research, New Delhi, India
3
National Institute of Biomedical Genomics, Kalyani, West Bengal, India
4
Department of Visceral Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Switzerland

Mini Review
Although tumor heterogeneity is widely accepted, the existence of cancer stem cells (CSCs) and their proposed role in tumor
maintenance has always been challenged and remains a matter of debate. Recently, a path-breaking chapter was added to
this saga when three independent groups reported the in vivo existence of CSCs in brain, skin and intestinal tumors using
lineage-tracing and thus strengthens the CSC concept; even though certain fundamental caveats are always associated with
lineage-tracing approach. In principle, the CSC hypothesis proposes that similar to normal stem cells, CSCs maintain self
renewal and multilineage differentiation property and are found at the central echelon of cellular hierarchy present within
tumors. However, these cells differ from their normal counterpart by maintaining their malignant potential, alteration of
genomic integrity, epigenetic identity and the expression of specific surface protein profiles. As CSCs are highly resistant to
chemotherapeutics, they are thought to be a crucial factor involved in tumor relapse and superficially appear as the ultimate
therapeutic target. However, even that is not the end; further complication is attributed by reports of bidirectional regenera-
tion mechanism for CSCs, one from their self-renewal capability and another from the recently proposed concept of dynamic
equilibrium between CSCs and non-CSCs via their interconversion. This phenomenon has currently added a new layer of com-
plexity in understanding the biology of tumor heterogeneity. In-spite of its associated controversies, this area has rapidly
emerged as the center of attention for researchers and clinicians, because of the conceptual framework it provides towards
devising new therapies.

The majority of tumors display substantial degree of pheno- ity to seed tumor in animal hosts and to divide in
typic and functional heterogeneity at the population level.1–3 asymmetric manner to allow self renewal as well as differen-
Heterogeneity among cancer cells arises within the same tiation into non-CSC progeny lacking tumor initiating capa-
tumor as a consequence of intrinsic, such as genetic4 and bilities (Fig. 1).8,9 Importantly, CSC rich tumors are
epigenetic changes5 as well as extrinsic factors like varied associated with aggressive disease, drug resistance and poor
microenvironment at different spatial locations.6 Tumor cell prognosis.10 CSC model advocates the presence of tumori-
heterogeneity may result from either clonal evolution driven genic stem-like cells in heterogeneous tumors, which differ-
by genetic instability and/or from differentiation of stem-like entiate into less-tumorigenic cancer cells to create a cellular
cells often called cancer stem cells (CSCs).7 The CSC model hierarchy.11 Moreover, the recently proposed concept of CSC
has rapidly emerged as the mainstream idea as it best fits plasticity suggests that these cell populations are dynamic
and logically explains the versatile attributes of heterogene- and both CSCs and non-CSCs are capable of interconversion
ous tumor populations. CSCs are characterized by their abil- in response to environmental cues, which further complicates
the situation in terms of generating tumor heterogeneity,
Key words: tumor heterogeneity, cancer stem cell, plasticity though the validity of this theory in in vivo is still controver-
*A.K.S., R.K.A. and S.M. contributed equally to this work sial.11–13 Despite the strong evidence supporting the CSC
Grant sponsor: CSIR (BSC106, BSC102, Fellowship Grants), MOES model in certain cancers, it is critical to acknowledge the
(GAP0118), UGC Fellowship Grant various limitations associated with CSC model in its concep-
DOI: 10.1002/ijc.28804 tual development such as reliability of cell surface marker
History: Received 8 Oct 2013; Accepted 11 Feb 2014; Online 22 Feb based identification of CSCs and lack of direct evidence
2014 about their in vivo existence.7 In recent times, reports com-
Correspondence to: Dipak Datta, Biochemistry Division, CSIR- bining mouse models that spontaneously generate tumors
CDRI, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, with genetic mapping put forward strong evidence in sup-
Lucknow-226031, India, Tel: 191-522-2772450 (Extn-4347/48), port of CSC concept.14–16 This article will not only highlight
Fax: 191-522-2771941, E-mail: dipak.datta@cdri.res.in the current controversies in CSC concept but also emphasize

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
1992 Tumor heterogeneity and CSC paradigm
Mini Review

Figure 1. Schematic overview of different models to explain cellular origin and maintenance of tumor along with their therapeutic interven-
tions and limitations. Stochastic model considers every cell within the tumor has equal tumorigenic potential. Classical CSC model assumes
stem-like cells are the key, which can irreversibly contribute to tumor heterogeneity by self-renewing as well as by their ability to differenti-
ate into other lineages. CSC plasticity model proposes bidirectional conversion equilibrium between CSCs and non-CSCs and emphasizes
the need for a lucrative combinatorial therapeutic approach to target CSCs as well as its biarmed regeneration mechanism.

the importance of tumor niche and various intracellular of cells called CSCs that are at the top of hierarchy, possess-
signaling cascades in shaping up tumor heterogeneity and ing self renewal and multilineage differentiation capabilities.8
CSC plasticity. There are four key characteristics that define the CSC popu-
lation: (a) compared with other tumor cells, only a small por-
CSC Hypothesis: Concept and Controversies tion of the cells within a tumor have tumorigenic potential
Concept when transplanted into immune-deficient mice; (b) it can be
In 1926, Bailey proposed that cancer was initiated and main- separated from other cancer cells based on their distinctive
tained by a small number of transformed precursor cells.17 In cell surface markers; (c) tumor, resulting from the CSCs, con-
late Nineties, Dick group in their pioneering studies, first tains mixed tumorigenic and nontumorigenic cells of the
documented and characterized the existence of CSCs in original tumor; and (d) it can be serially transplanted
hematopoietic malignancies.18 Later, the CSC concept has through multiple generations, indicating its self renewing
been extensively expanded to cover most of the solid capabilities.21 It is noteworthy to mention that all cancers do
tumors.19,20 As per the CSC model, similar to their tissue of not necessarily follow CSC model and in such cancers,
origin, tumor cells are organized hierarchically with a subset although genetic and/or phenotypic heterogeneity are present,

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
Singh et al. 1993

they are equally tumorigenic and do not possess cellular hier- studies are the CSCs but such tumorigenic potential can only
archy in them.22,23 It has been also proposed that clonal evo- be tested in a permissive environment. Sceptics have persis-
lution model and CSC model may act together in a tently pointed out that transplantation removes cells from
cooperative manner within the same tumor to drive tumor their native environment and may therefore, change their
cell heterogeneity.24 In light of new reports about reversibility behavior and tumorigenic potential.33 Moreover, transplanta-
of cancer cells,11–13 stochastic and CSC models are inter- tion studies do not provide any scope to test whether human
linked and CSC model is shifting from a rigid hierarchical to cells have tumorigenic potential if they are killed by the xen-
a more flexible model to encompass a larger group of tumors ogeneic immune response. Simply demonstrating the in vivo
within its domain.25 tumorigenic and nontumorigenic capabilities of a tumor cell
to an immune-compromised animal host is not sufficient for
Controversies the CSC model and other criteria like the potential of

Mini Review
Central to the controversy over the CSC concept is the lack tumorigenic cells to give rise to non-tumorigenic progeny has
of benchmark functional assays, over-reliance on cell surface to be satisfied. Without demonstrating a fate map for lineage
markers, inconsistencies in terminology and plasticity relationship between CSCs and non-CSCs, the functional dif-
between cell types. Especially in case of solid tumors, the ferences between these cells may be considered as a result of
identification of CSCs by so called defined cell surface genetic differences. There are several technical challenges
markers has poor reproducibility and our tendency not to associated with the study of CSC fate in vivo; such as diffi-
publish data that are difficult to interpret or inconsistent culty in distinguishing them from their differentiated prog-
with existing model has amplified the perplexity of our eny, scarcity of surface markers exclusively present on CSCs
understanding.26,27 As an example, initially CD133 was pro- and specifically associated with its phenotype. However, with
posed to robustly distinguish CSCs from non-CSCs in brain28 the technical advancement, combining classical lineage-
and colon29 cancer cells but a series of subsequent studies tracing approach with marker based assays, three different
found that CD1332 also have similar tumorigenic potential groups independently demonstrated the fate of CSCs in vivo
in vivo.30–32 Additionally, markers expressed by CSCs differ in tumor progression.14–16 Driessens et al., in their elegant
among patients suffering from the same type of cancer. A studies found that benign papillomas are hierarchically
key question raised by such differences is whether tumors of organized and followed CSC model in vivo.14 Employing fate
the same type differ in the extent to which they are hierarchi- map by selective ablation of Nestin1 CSCs in mouse model
cally organized.27 Another important parameter is to judge of glioma, Chen et al. demonstrated that Nestin1 cells are
the tumorigenic potential of CSCs under in vivo condition main driving force for tumor growth, therapy resistance and
where it largely relies on the immune-competency potential tumor relapse.15 However, long term effects of selective abla-
of the host. At least, in the case of melanoma, Quintana et al. tions of Nestin1 cells could not be assessed as mice inde-
convincingly proved that modification of xenotransplantation pendently developed lethal tumors other than the original
assays could dramatically increase the detectable frequency of ones. Therefore, the exact contribution of Nestin1 cells
tumorigenic cells that are commonly known as a rare popula- remains unclear in terms of driving long lasting tumor
tion.23 Further controversy in CSC model exists at the level growth and its possible interrelationship with Nestin2 cells.
of origin and terminology of CSCs during tumor initiation. By using multicolour conditional reporter assay, Schepers
The term “CSC”’ could be misleading, when it implies that et al. discovered that in accordance with CSC theory, most of
such cells can only be derived from the stem cells of the cor- the Lgr51 intestinal CSCs differentiated into Lgr52 non-
responding tissue.21 CSCs may come up from normal stem CSCs in vivo, though Lgr52 cells were not being fate mapped
cells by mutations of specific oncogenes that make them can- in their experiment.16 Interestingly, in contrast to the previ-
cerous, but again this may not be the case in all tumors. In ous finding, a recent study shows that Lgr52 cells can also
respect to the recently proposed CSC plasticity in solid act as cell of origin for intestinal adenomas and seems not to
tumors, CSC may even arise from cells that are of non stem follow the CSC model.34 This counter challenging evidence
cell, obviously put forward their stemness identity in jeop- may possibly support the concept of CSC plasticity where
ardy.25 Therefore, careful optimisation of assay system and non-CSCs can be dedifferentiated into CSCs since Lgr52 cells
using multiple combinations of endogenous and/or surface can give rise to Lgr51 cells in in vivo condition.
markers will possibly provide a clearer picture as well help us These reports provided a strong support about the pres-
to identify, isolate and characterise quintessential CSCs in ence of stem-like cells in heterogeneous tumor populations in
particular cancer types. vivo and propelled the use of modern form of lineage tracing
in CSC biology; nevertheless it did not end the CSC debate
In Vivo CSC Existence: Is It Resolving the Debate? as lineage tracing has its own limitations in noisy and
Until recently, most of the CSC literature addressed the poten- dynamic tumor microenvironment.35–38 Most of the lineages
tial of these cells to contribute to the disease, but not the tracing approaches use DNA-label retention or inducible
actual fate of the individual cell in real in vivo situation. In genetic labelling and follow the fate of labelled cells and their
principle, the cells that generate new tumors in transplantation progenies using single or multicolour reporters depending on

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
1994 Tumor heterogeneity and CSC paradigm

single or multiple markers used. During lineage tracing, it is a part of cancer cell plasticity but all cancer cell plasticity do
imperative to note that DNA-label retention is not only not belong to CSC plasticity.
restricted to quiescent stem-like cells, but also a characteristic Although, the underlying molecular mechanism for CSC
of post-mitotic highly differentiated senescent cells. Also, it is plasticity is yet unknown, conceptually it links clonal evolu-
of utmost importance to ensure that the site of tracing initia- tion and CSC models and demands that CSC hierarchy must
tion should be carefully mapped as recombinant alleles or be seen as bidirectional balance between CSCs and differenti-
transgenes are often mosaically expressed between different ated non-CSCs. Whether, CSC plasticity is orchestrated by
group of cells.36 Another constraint of lineage tracing using microenvironmental cues or simply a stochastic process may
cell transplantation is that single cells may not behave in the depend on the type of tumor and its genetic background.25
context of a graft as they do during normal tissue homeosta- Although lineage tracing studies provided strong evidence in
sis as lineage tracing in tumor epithelial cells demonstrated favor of the presence and differentiation of CSCs in vivo in
Mini Review

altered differentiation pattern compared to intact tissue.37 For different solid tumors, they did not exclude the possibility
example, lineage tracing in mammary and epidermal cell that labelled CSCs might itself arise from more differentiated
transplantation studies have shown that the labelled cells non-CSCs. It is also important to note that nearly all existing
acquire multipotent phenotype though this is not observed in reports about reversible transition between tumorigenic
the event of lineage tracing in undamaged tissues.39 High (CSCs) and nontumorigenic (non-CSCs) states come from
coverage sequencing of human tumors as well as live imaging studies on cell lines. As certain high passage cell lines have
of human tumors is emerging as alternative tools to study been found to show endogenous phenotypic plasticity, it
the fate choice of tumor cells in driving tumor heterogeneity, would be critical to overcome this possibility by investigating
although their feasibility is yet to be tested.38 the extent of such plasticity in spontaneously arising tumors
in vivo.27 In support of in vivo CSC plasticity, Verma and
Solid Tumor Heterogeneity and CSC Plasticity coworkers reported that glioma can be generated from termi-
Within a solid tumor, a remarkable variability is found in the nally differentiated neurons which can revert to a stem-like
morphological and physiological feature of tumor cell popu- state and express typical CSC markers following transforma-
lation. Although cancer cell heterogeneity was classically pro- tion, highlighting the pivotal role of genetic perturbations in
posed after the seminal studies of Josh Fidler, Gloria influencing CSC plasticity.44 More in depth in vivo lineage
Heppner and others in the seventies,40,41 experimental explo- tracing with multicolour labelling of non-CSCs and CSCs
ration of this subject had been limited by the conceptual may elucidate differentiation hierarchies and CSC plasticity
framework as well as by the unavailability of relevant study in future.
tools. However, the discovery that tumor formation is
dependent on the acquisition of oncogenic mutations Factors Governing Solid Tumor Heterogeneity and
changed the situation. These findings underlined the gene CSC Plasticity
centric prospective of tumor heterogeneity and is thought to The origin of intra-tumor heterogeneity and initial CSC gen-
be attributed by genetic diversity resulting from clonal evolu- eration within a tumor are still a matter of debate and
tion. Moreover, dominance of gene centric views has been remains an enigma to researchers. There are various factors
challenged by the CSC hypothesis which additionally brings that have significant role in maintaining CSC plasticity as
non-genetic sources of phenotypic variability into the focus. well as solid tumor heterogeneity, but their relative contribu-
As a result of this, in recent years, tumor heterogeneity is tion to each of these phenomena is not clear yet. It seems
viewed under the glasses encompassing its genetic, epigenetic plausible that all the coordinated actions of these factors are
and phenotypic heterogeneity.42 necessary to shape the phenotypic heterogeneity and govern
Certain cancer cells reversibly fluctuate among the states CSC plasticity.
with varying degree of competence to contribute to tumor
growth.11,43 An attractive model to explain this differential Tumor microenvironment and CSC niche
ability of tumor cells is the newly established concept of The microenvironment within a solid tumor is not at all
“CSC plasticity” under which the majority of tumor cells can homogeneous; rather it is a very complex network of versatile
be considered as stem cells with varying degree of “stemness” factors and among the key forces in driving tumor heteroge-
governed by microenvironmental cues and other stochastic neity.45 Unique combination of growth factors secreted by
cell autonomous mechanisms (Fig. 1).12 Although lot of scep- tumor residing cells play a pivotal role in differentiating
ticism persist in the terminology of tumor cell plasticity in CSCs and non-CSCs and vice versa and ultimately generates
the published literature, CSC plasticity will be more appropri- complex heterogeneous tumors. Variations are also found at
ate as it includes at least one or both stem-like cells out of the level of lymphatic vasculature, diverse oxygen supply, dif-
two inter-converting cells. Cancer cell plasticity would better ferent numbers and types of stromal cells, infiltrating cells
fit between the inter-converting tumor cells with less tumori- and various composition of extracellular matrix along with
genic potential and differing from each other by phenotypes diverse physico-chemical properties. Therefore, cancer cells
that are independent of stemness. In theory, CSC plasticity is within a given tumor are expected to experience a wide range

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
Singh et al. 1995

of micro-environmental cues, as for example, well oxygenated development, progression, metastasis, drug resistance and
tumor periphery versus hypoxic tumor core environment, even in CSC maintenance and plasticity.57 Yu et al. were the
which would in turn translate into a series of phenotypic first to study miRNA expression in breast CSCs where they
changes including tumor heterogeneity and CSC plasticity.42 observed significant downregulation of let-7, miR-16, miR-
CSCs are believed to be frequently located at the hypoxic 107, miR-128 and miR-20b in CD441/highCD242/low cells
core of solid tumors and by their unique adaptability to that compared to non-CSCs.58 Seminal studies by the Struhl
particular environment, pose a serious threat to the antitu- group have put forward the direct evidence for the role of
mor effects of chemotherapeutic drugs. CSCs are not only miRNA(s) in regulating CSC plasticity in breast cancer
intrinsically programmed to fulfil their detrimental roles, but patients where the expression of miR-200 family members
are also orchestrated by stromal cells residing in their vicinity are selectively downregulated during the acquisition of CSC
in forming a CSC niche.46 As for example, IFNg inducible phenotype from non-CSCs.59,60 Indeed, miR-200b and miR-

Mini Review
chemokines and CXCL12 secreted by tumor infiltrating and 200c overexpression strongly inhibited the proliferation of
stromal cells interact with their receptors CXCR3, CXCR4 CSCs and their ability to form tumors and this effect was
(pancreatic and prostate CSC marker) and CXCR7 and sub- mediated by targeting different subunits of the polycomb
sequent signaling crosstalk may result in amplification of the group (PcG) of proteins (Fig. 2).59–61 Stem cells rely on PcG
CSC loop.47,48 Interactions between CSCs and tumor endo- proteins to reversibly repress genes encoding transcription
thelial cells also play a major role in sequestering the angio- factors required for differentiation.62 Various groups
genic process as documented in brain cancer where Nestin1/ hypothesize that the acquisition of promoter DNA methyla-
CD1331 CSCs are located in direct proximity of endothelial tion at these repressed genes could lock in stem cell pheno-
cells. Considering the close association between CSCs and type to initiate abnormal clonal expansion and thereby
tumor endothelial cells, it has been proposed that newly predispose to cancer.63 Furthermore, CDKN2A (p16) and
formed vasculature within a growing tumor can be derived CDKN2D (p19) have been found to be transcriptionally
from the differentiation of CSCs implicating its multilineage repressed by PcG proteins in human embryonic stem cells
differentiation capabilities.49,50 and form polycomb repressive complex 2 (PRC2), which are
associated with nucleosomes that are trimethylated at Lys27
Genetic instability/mutation of histone H3 (H3K27).64 It has been observed that in case
Although genetic instability is widely considered as a hall- of various cancers including breast, prostate, ovary and pan-
mark of cancer, its precise role in CSC generation and plas- creas, the isolated CSC population overexpresses enhancer of
ticity is still uncertain. The notion that CSCs can recapitulate zeste homolog 2 (EZH2), one of the key members of PRC2,
the heterogeneity of the original tumor in transplantation responsible for trimethylating H3K27, which is essential for
studies is based on certain surface markers only. At least, at maintenance of an intact CSC population.65,66 Particularly,
the level of initiation, spontaneous tumors are not similar to PRC1 acts downstream of PRC2 and recognizes the repres-
experimental tumors induced by transplantation or by few sive H3K27me3 marks and inhibits the transcription of genes,
defined genetic mutations; rather it arises from a much more responsible for stem cell differentiation. This effect of PRC1
complex process of somatic evolution. Acquisition of neces- in postponing the commitment of stem cells has been found
sary driving mutations is always associated with a larger to be mediated by its BMI1 (B cell specific Moloney murine
number of genetic alterations, which are usually silent in leukemia virus integration site 1) component via interacting
nature.42 Hence, not every genetic difference between the with Ring1b.67 Furthermore, BMI1 and EZH2 are overex-
cells contributes to phenotypic variability but mutations that pressed in CSCs and found to be necessary for tumor initia-
are normally silent might increase the phenotypic plasticity tion of various solid tumors such as head and neck cancer
towards stemness and drug resistant stage via differential and glioblastoma.68 Recently, it has been reported that TGF-
gene expression. As an example, multidrug resistance pro- b enhances CSC plasticity by reducing the presence of
teins such as ABCG2 are often mutated in their substrate H3K27me3 repressive histone modification at the ZEB1 (zinc-
binding pocket enhancing their drug resistance effects and finger E box-binding homeobox 1) promoter site and induce
favoring CSC like properties.51 Data from AML, breast, renal non-CSC to CSC conversion in breast cancer.69,70 miR-200c
and pancreatic cancer show extensive genetic heterogeneity repressed the expression of BMI1, while loss of miR-200b
which would not be driven by merely differentiation of CSC increased suppressor of zeste 12 homolog (SUZ12) expression
but rather every CSC could form genetically distinct tumor and H3K27 methylation.61 Interestingly, ectopic expression of
population.52–56 Extensive genetic heterogeneity also provides SUZ12 in transformed cells was able to generate CSCs.60 In
opportunity for these genetic changes to confer phenotypic addition, miR-200c levels are regulated by a complicated loop
and functional heterogeneity among tumors. comprising BMI1 and ZEB1.71 These findings revealed that
miR-200 family members play an important role in regula-
MicroRNAs and other epigenetic factors tion of CSC formation and function via PcG complexes.
In the last few years, multiple miRNAs have been reported as miR-200 is also involved in modulating the expression of
modulators of transcription factors involved in tumor SRY (sex determining region Y)-box 2 (SOX2), Kruppel-like

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
1996 Tumor heterogeneity and CSC paradigm
Mini Review

Figure 2. Schematic illustration of epigenetic regulation of CSC plasticity via polycomb repressor complexes (PRCs). Sequential events
describe miR-200 family mediated reversible regulation of PRC1 and PRC2 formation to maintain closed and open chromatin state in CSCs
and non-CSCs, respectively, to subsequently control the expression of differentiation phenotypes via p16 and p19 proteins.

factor 4 (KLF4), BMI1 and SUZ12 genes, previously described (JAK1)—Signal transducer and activator of transcription 3
to regulate stemness in cancer cells.61,72 Furthermore, (STAT-3)/IL-6-JAK1-STAT3, Epidermal growth factor recep-
through unbiased miRNA expression profiling, Tang and col- tor (EGFR) signaling and their crosstalk with stemness
leagues recently showed that prostate CSCs selectively down- related transcription factors play a pivotal role in regulating
regulate miR-34a and let-7b. In addition, miR-34a was shown CSC plasticity in solid tumors (Fig. 3).13,59,74
to directly target stem cell related genes like CD44 and
NOTCH, implying a crucial role of miRNAs in maintaining Classical stem cell signaling (Wnt, notch and hedgehog)
the CSC phenotype by halting their differentiation.57 and crosstalk in CSC plasticity
Wnt/b-catenin, Notch and Hedgehog signaling pathways are
Signaling Events Involved in CSC Maintenance and highly versatile, complex and found in embryonic or adult
Plasticity stem cells in its normal form as well as in CSCs in subverted
Although, CSCs are unique in their ability to generate form.75 Interestingly, many of the cell surface markers such
tumors, they also share various similarities to normal adult as Lgr5, CD44, CD24 and CD326, which have been used to
tissue stem cells. CSCs often inappropriately use many signal- identify putative CSCs are Wnt targets suggesting its direct
ing pathways used by their normal counterpart and present a role in the acquisition of CSC phenotype.73,76,77 Wnt/b-cate-
challenge to the development of CSC specific therapies.73 nin signaling transcriptionally regulates the expression of
Besides involving classical stem cell signaling pathways, it has OCT4, which is the key controller of CSC self renewal and
also been shown that interleukin-6 (IL-6)—Janus Kinase 1 maintenance of stemness (Fig. 3). Wnt/b-catenin itself or by

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
Singh et al. 1997

Mini Review
Figure 3. Schematic representation of signaling crosstalk in CSC plasticity. Classical stem cell specific (Wnt, Hedgehog and Notch) and
newly discovered (IL-6, EGFR and HIF) signaling pathways and their crosstalk in CSC maintenance and plasticity. BMI1 may act as a major
regulator of CSC state and intermediate protein linking various pathways, making it a potential target for future chemotherapeutic
strategies.

interacting with other downstream partners of Hedgehog sig- of CSC phenotype either by inhibiting p16 and p19 transcrip-
naling pathway, promotes the expression of Jagged1/Jagged2 tion or by other unidentified functions.85
that are known ligands for Notch pathway.78 Hedgehog sig-
naling has been shown to regenerate CSCs by inducing epi- Novel role of IL-6 and EGF signaling in CSC plasticity
thelial to mesenchymal transition (EMT) via up-regulation of Although CSC plasticity is a reversible phenomenon, the rate
the transcription factor SNAIL and the concomitant down of interconversion between CSCs and non-CSCs differ signifi-
regulation of E-cadherin.79 It has been demonstrated that cantly. It has been demonstrated that CSCs rapidly differenti-
Hedgehog stimulation in culture increased the number and ate into non-CSCs, but non-CSCs are not easily convertible
size of mammospheres whereas, cyclopamine (a Hedgehog into CSCs. This nonreciprocal conversion process between
pathway inhibitor) had the opposite effect indicating the non-CSCs and CSCs should result in the ever decreasing
direct relationship between Hedgehog pathway and CSC gen- number of CSCs, which is paradoxical, as CSCs are main-
eration.80 The transcriptional program orchestrated by tained at a constant proportion within a tumor.13 This obser-
Hedgehog signaling depends on the Gli family of transcrip- vation led to the foundation that CSCs and non-CSCs
tion factors.81 Although Gli proteins can be converted to interact with each other in a paracrine fashion and maintain
either transcriptional activators or truncated transcriptional a dynamic equilibrium in a heterogeneous tumor population
repressors, Hedgehog-Gli interaction ultimately enhances self that do not occur when cells are cultured separately. In the
renewal properties and CSC plasticity either by the direct search of paracrine factors, it has recently been shown that
binding of Gli activator (GliA) to the promoters of SNAIL, IL-6 plays a crucial role in maintaining CSC plasticity. As its
CYCLIN D, MYC and JAGGED2 etc. or via activating the key downstream target, IL-6 regulates CSC-associated OCT4,
stemness regulator BMI1 through GliR (Fig. 3).82–84 BMI1 SOX2 and NANOG gene expressions through the IL-6-JAK1-
plays a central role in the crosstalk of classical stem cell path- STAT3 signal transduction pathway (Fig. 3), which converts
ways and finally results in the maintenance and acquisition non-CSCs into CSCs.74 Transcription factor STAT3 is

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
1998 Tumor heterogeneity and CSC paradigm

activated through phosphorylation of tyrosine residue 705 development of new therapeutic strategies considering CSCs
and results in nuclear translocation and activation of the and its relationship with tumor microenvironment are
STAT3 transcriptional regulatory function.86,87 Interestingly, urgently required as tumor milieu is believed to be the key
STAT3 activation is mediated by members of the JAK family source for cancer cell differentiation and bidirectional conver-
proteins as well as receptor tyrosine kinase (RTK) family pro- sion.25,94 Therefore, identification of factors responsible for
teins via EGFR.88 EGF-RTK signaling pathway plays a critical bidirectional conversion of tumor cells will be of fundamental
role in regulating CSC state via its multiarmed downstream importance.95 Keeping the newly discovered concept of CSC
cascades and acts as a link for crosstalk between Hedgehog plasticity in mind, effective cancer therapy should not only
and IL-6 signaling. PI3K activated via EGFR signaling can target CSCs but also prevent CSC generation from non-CSCs
either turn on GliA via PI3K-AKT-mTORC1-S6K pathway to (Fig. 1) as CSCs can be recreated from bulk non-CSC popu-
induce the expression of CSC specific genes or ligands for lation at any stage of tumorigenesis. In this respect, the lead-
Mini Review

other pathways (Jagged 2),89 or it can activate STAT3 via ing candidate drug is the ionophore antibiotic salinomycin
Bone marrow X-linked protein to express the stemness spe- and a well known type 2 diabetic drug metformin, which
cific genes like OCT4, SOX2, CD44, etc (Fig. 3).90,91 Studies have recently been documented to successfully eliminate
have suggested that the core hypoxic zone of solid tumors CSCs in different types of human cancers in vitro and in vivo
harbors highly resistant tumor cells overexpressing CSC and are also being highly effective while used in combina-
markers. Hypoxic condition within a tumor can induce stem- tions with other conventional chemotherapeutic drugs.96–101
ness in tumor population via transcriptional induction of Recently, pharmacological inhibitors of S phase kinase associ-
hypoxia inducible factor (HIF)1a and HIF2a.92 HIF1a can ated protein 2 (SKP2) have been demonstrated to effectively
also be activated via PI3K-AKT to induce EMT modulator inhibit CSCs and considered for their therapeutic poten-
Twist1 and polycomb repressor BMI1 to help the downregu- tial.102,103 However, as SKP2 is also involved in general
lation of p16 and p19 that are required for the differentiation metabolism, clinical efficacy and toxicological concerns of its
of CSCs. Unlike HIF1a, HIF2a acts as a transcriptional acti- inhibitors need to be further tested. In cancers that do not
vator of stemness specific genes OCT4, SOX2, CD44, KLF-4, follow the CSC model, it would be sensible to avoid unusual
MYC, etc.93 Hence, it is noteworthy that these newly discov- highlighting of the CSC theory towards drug development by
ered signaling pathways act in synergistic manner with an focusing on small population of cancer cells as they do not
overall goal of inducing stemness and further elucidation of have specific capacity to drive disease progression or tumor
their complex crosstalk will help in better understanding of relapse. Further investigation of molecular mechanism of
CSC plasticity. CSC plasticity, understanding the mode of action of CSC spe-
cific drugs and their clinical efficacy are required to offer
Therapeutic Potential and Concluding Remarks highly effective therapies for patients in all stages of cancer.
Although intermittent improvements are being made, tumor
relapse and treatment resistance remains a major cause of
morbidity and mortality that are proposed to be particularly
Acknowledgements
The authors are grateful to Dr. David M Briscoe for reading the manuscript
due to the persistence of highly resistant CSC population and his thoughtful insight. Institutional (CSIR-CDRI) communication num-
during chemotherapy at least in the case of cancers following ber for this article is 8623. DD dedicates this manuscript to his beloved
CSC model. Hence, for complete eradication of cancer cells, teacher Dr. Samar Chakraborty.

References
1. Fidler IJ, Hart IR. Biological diversity in meta- 8. Clarke MF, Dick JE, Dirks PB, et al. Cancer cells via IL6 secretion. Proc Natl Acad Sci USA
static neoplasms: origins and implications. Sci- stem cells-perspectives on current status and 2011;108:1397–402.
ence 1982;217:998–1003. future directions: AACR Workshop on cancer 14. Driessens G, Beck B, Caauwe A, et al. Defining
2. Heppner GH. Tumor heterogeneity. Cancer Res stem cells. Cancer Res 2006;66:9339–44. the mode of tumour growth by clonal analysis.
1984;44:2259–65. 9. Tang DG. Understanding cancer stem cell heter- Nature 2012;488:527–30.
3. Marusyk A, Polyak K. Tumor heterogeneity: ogeneity and plasticity. Cell Res 2012;22:457–72. 15. Chen J, Li Y, Yu TS, et al. A restricted cell pop-
causes and consequences. Biochim Biophys Acta 10. Dean M, Fojo T, Bates S. Tumour stem cells ulation propagates glioblastoma growth after
2010;1805:105–17. and drug resistance. Nat Rev Cancer 2005;5: chemotherapy. Nature 2012;488:522–6.
4. Nowell PC. The clonal evolution of tumor cell 275–84. 16. Schepers AG, Snippert HJ, Stange DE, et al. Lin-
populations. Science 1976;194:23–8. 11. Gupta PB, Fillmore CM, Jiang G, et al. Stochas- eage tracing reveals Lgr51 stem cell activity in
5. Baylin SB, Jones PA. A decade of exploring the tic state transitions give rise to phenotypic equi- mouse intestinal adenomas. Science 2012;337:
cancer epigenome - biological and translational librium in populations of cancer cells. Cell 2011; 730–5.
implications. Nat Rev Cancer 2011;11:726–34. 146:633–44. 17. Bailey H. The abdominal crises of pernicious
6. Polyak K, Weinberg RA. Transitions between 12. Chaffer CL, Brueckmann I, Scheel C, et al. Nor- anaemia. Br Med J 1926;2:554.
epithelial and mesenchymal states: acquisition of mal and neoplastic nonstem cells can spontane- 18. Lapidot T, Sirard C, Vormoor J, et al. A cell ini-
malignant and stem cell traits. Nat Rev Cancer ously convert to a stem-like state. Proc Natl tiating human acute myeloid leukaemia after
2009;9:265–73. Acad Sci USA 2011;108:7950–5. transplantation into SCID mice. Nature 1994;
7. Shackleton M, Quintana E, Fearon ER, et al. 13. Iliopoulos D, Hirsch HA, Wang G, et al. Induci- 367:645–8.
Heterogeneity in cancer: cancer stem cells versus ble formation of breast cancer stem cells and 19. Al-Hajj M, Wicha MS, Benito-Hernandez A,
clonal evolution. Cell 2009;138:822–9. their dynamic equilibrium with non-stem cancer et al. Prospective identification of tumorigenic

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
Singh et al. 1999

breast cancer cells. Proc Natl Acad Sci USA between two distinct phenotypes that are prefer- stem cells with normal stem cells. Cell 2009;138:
2003;100:3983–8. entially migratory or proliferative. Cancer Res 592–603.
20. Visvader JE, Lindeman GJ. Cancer stem cells in 2011;71:5317–26. 62. Ringrose L, Paro R. Epigenetic regulation of cel-
solid tumours: accumulating evidence and unre- 44. Friedmann-Morvinski D, Bushong EA, Ke E, lular memory by the Polycomb and Trithorax
solved questions. Nat Rev Cancer 2008;8:755–68. et al. Dedifferentiation of neurons and astrocytes group proteins. Annu Rev Genet 2004;38:413–
21. Gupta PB, Chaffer CL, Weinberg RA. Cancer by oncogenes can induce gliomas in mice. Sci- 43.
stem cells: mirage or reality? Nat Med 2009;15: ence 2012;338:1080–4. 63. Schuebel K, Chen W, Baylin SB. CIMPle origin
1010–2. 45. Medema JP, Vermeulen L. Microenvironmental for promoter hypermethylation in colorectal
22. Kelly PN, Dakic A, Adams JM, et al. Tumor regulation of stem cells in intestinal homeostasis cancer? Nat Genet 2006;38:738–40.
growth need not be driven by rare cancer stem and cancer. Nature 2011;474:318–26. 64. Lee TI, Jenner RG, Boyer LA, et al. Control of
cells. Science 2007;317:337. 46. Fessler E, Dijkgraaf FE, De Sousa E, et al. Can- developmental regulators by Polycomb in
23. Quintana E, Shackleton M, Sabel MS, et al. Effi- cer stem cell dynamics in tumor progression human embryonic stem cells. Cell 2006;125:301–
cient tumour formation by single human mela- and metastasis: is the microenvironment to 13.
noma cells. Nature 2008;456:593–8. blame? Cancer Lett 2013;341:97–104. 65. Crea F, Hurt EM, Mathews LA, et al. Pharmaco-

Mini Review
24. Anderson K, Lutz C, van Delft FW, et al. 47. Singh AK, Arya RK, Trivedi AK, et al. Chemo- logic disruption of polycomb repressive complex
Genetic variegation of clonal architecture and kine receptor trio: CXCR3, CXCR4 and CXCR7 2 inhibits tumorigenicity and tumor progression
propagating cells in leukaemia. Nature 2011;469: crosstalk via CXCL11 and CXCL12. Cytokine in prostate cancer. Mol Cancer 2011;10:40.
356–61. Growth Factor Rev 2013;24:41–9. 66. Rizzo S, Hersey JM, Mellor P, et al. Ovarian
25. Medema JP. Cancer stem cells: the challenges 48. Datta D, Flaxenburg JA, Laxmanan S, et al. Ras- cancer stem cell-like side populations are
ahead. Nat Cell Biol 2013;15:338–44. induced modulation of CXCL10 and its receptor enriched following chemotherapy and overex-
26. Lathia JD. Cancer stem cells: moving past the splice variant CXCR3-B in MDA-MB-435 and press EZH2. Mol Cancer Ther 2011;10:325–35.
controversy. CNS Oncol 2013;2:465–7. MCF-7 cells: relevance for the development of 67. Pietersen AM, van Lohuizen M. Stem cell regu-
27. Meacham CE, Morrison SJ. Tumour heterogene- human breast cancer. Cancer Res 2006;66:9509– lation by polycomb repressors: postponing com-
ity and cancer cell plasticity. Nature 2013;501: 18. mitment. Curr Opin Cell Biol 2008;20:201–7.
328–37. 49. Jain RK, Carmeliet P. SnapShot: tumor angio- 68. Chang CJ, Hung MC. The role of EZH2 in
28. Singh SK, Hawkins C, Clarke ID, et al. Identifi- genesis. Cell 2012;149:1408–e1. tumour progression. Br J Cancer 2012;106:243–
cation of human brain tumour initiating cells. 50. Jiang B, Mason J, Jewett A, et al. Tube-like 7.
Nature 2004;432:396–401. structures with co-expression of D2–40 and 69. Chaffer CL, Marjanovic ND, Lee T, et al. Poised
29. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. CD34: newly formed vasculatures? Int J Biol Sci chromatin at the ZEB1 promoter enables breast
Identification and expansion of human colon- 2012;8:1206–16. cancer cell plasticity and enhances tumorigenic-
cancer-initiating cells. Nature 2007;445:111–5. 51. Zoernig I, Ziegelmeier C, Lahrmann B, et al. ity. Cell 2013;154:61–74.
30. Wang J, Sakariassen P, Tsinkalovsky O, et al. Sequence mutations of the substrate binding 70. Marjanovic ND, Weinberg RA, Chaffer CL.
CD133 negative glioma cells form tumors in pocket of stem cell factor and multidrug resist- Poised with purpose: cell plasticity enhances
nude rats and give rise to CD133 positive cells. ance protein ABCG2 in renal cell cancer: a pos- tumorigenicity. Cell Cycle 2013;12:2713–4.
Int J Cancer 2008;122:761–8. sible link to treatment resistance. Oncol Rep 71. Wellner U, Schubert J, Burk UC, et al. The
31. Chen R, Nishimura MC, Bumbaca SM, et al. A 2013;29:1697–700. EMT-activator ZEB1 promotes tumorigenicity
hierarchy of self-renewing tumor-initiating cell 52. Ding L, Ley TJ, Larson DE, et al. Clonal evolu- by repressing stemness-inhibiting microRNAs.
types in glioblastoma. Cancer Cell 2010;17:362–75. tion in relapsed acute myeloid leukaemia Nat Cell Biol 2009;11:1487–95.
32. Joo KM, Kim SY, Jin X, et al. Clinical and bio- revealed by whole-genome sequencing. Nature 72. Gregory PA, Bert AG, Paterson EL, et al. The
logical implications of CD133-positive and 2012;481:506–10. miR-200 family and miR-205 regulate epithelial
CD133-negative cells in glioblastomas. Lab 53. Nik-Zainal S, Van Loo P, Wedge DC, et al. The to mesenchymal transition by targeting ZEB1
Invest 2008;88:808–15. life history of 21 breast cancers. Cell 2012;149: and SIP1. Nat Cell Biol 2008;10:593–601.
33. Gilbertson RJ, Graham TA. Cancer: resolving 994–1007. 73. LaBarge MA. The difficulty of targeting cancer
the stem-cell debate. Nature 2012;488:462–3. 54. Xu X, Hou Y, Yin X, et al. Single-cell exome stem cell niches. Clin Cancer Res 2010;16:3121–
34. Schwitalla S, Fingerle AA, Cammareri P, et al. sequencing reveals single-nucleotide mutation 9.
Intestinal tumorigenesis initiated by dedifferenti- characteristics of a kidney tumor. Cell 2012;148: 74. Kim SY, Kang JW, Song X, et al. Role of the IL-
ation and acquisition of stem-cell-like properties. 886–95. 6-JAK1-STAT3-Oct-4 pathway in the conversion
Cell 2013;152:25–38. 55. Campbell PJ, Yachida S, Mudie LJ, et al. The of non-stem cancer cells into cancer stem-like
35. Kretzschmar K, Watt FM. Lineage tracing. Cell patterns and dynamics of genomic instability in cells. Cell Signal 2013;25:961–9.
2012;148:33–45. metastatic pancreatic cancer. Nature 2010;467: 75. Holland JD, Klaus A, Garratt AN, et al. Wnt
36. Barker N, van Oudenaarden A, Clevers H. Iden- 1109–13. signaling in stem and cancer stem cells. Curr
tifying the stem cell of the intestinal crypt: strat- 56. Gerlinger M, Rowan AJ, Horswell S, et al. Intra- Opin Cell Biol 2013;25:254–64.
egies and pitfalls. Cell Stem Cell 2012;11:452–60. tumor heterogeneity and branched evolution 76. Liu BY, McDermott SP, Khwaja SS, et al. The
37. Watt FM, Jensen KB. Epidermal stem cell diver- revealed by multiregion sequencing. N Engl J transforming activity of Wnt effectors correlates
sity and quiescence. EMBO Mol Med 2009;1: Med 2012;366:883–92. with their ability to induce the accumulation of
260–7. 57. Liu C, Kelnar K, Liu B, et al. The microRNA mammary progenitor cells. Proc Natl Acad Sci
38. Blanpain C, Simons BD. Unravelling stem cell miR-34a inhibits prostate cancer stem cells and USA 2004;101:4158–63.
dynamics by lineage tracing. Nat Rev Mol Cell metastasis by directly repressing CD44. Nat Med 77. Takahashi-Yanaga F, Kahn M. Targeting Wnt
Biol 2013;14:489–502. 2011;17:211–5. signaling: can we safely eradicate cancer stem
39. Van Keymeulen A, Rocha AS, Ousset M, et al. 58. Yu F, Yao H, Zhu P, et al. let-7 regulates self cells? Clin Cancer Res 2010;16:3153–62.
Distinct stem cells contribute to mammary renewal and tumorigenicity of breast cancer 78. Takebe N, Ivy SP. Controversies in cancer stem
gland development and maintenance. Nature cells. Cell 2007;131:1109–23. cells: targeting embryonic signaling pathways.
2011;479:189–93. 59. Polytarchou C, Iliopoulos D, Struhl K. An inte- Clin Cancer Res 2010;16:3106–12.
40. Fidler IJ. Tumor heterogeneity and the biology grated transcriptional regulatory circuit that 79. Chou CH, Lieu AS, Wu CH, et al. Differential
of cancer invasion and metastasis. Cancer Res reinforces the breast cancer stem cell state. Proc expression of hedgehog signaling components
1978;38:2651–60. Natl Acad Sci USA 2012;109:14470–5. and Snail/E-cadherin in human brain tumors.
41. Heppner GH. Tumor heterogeneity. Cancer Res 60. Iliopoulos D, Lindahl-Allen M, Polytarchou C, Oncol Rep 2010;24:1225–32.
1984;44:2259–65. et al. Loss of miR-200 inhibition of Suz12 leads 80. Rubin LL, de Sauvage FJ. Targeting the Hedge-
42. Marusyk A, Almendro V, Polyak K. Intra- to polycomb-mediated repression required for hog pathway in cancer. Nat Rev Drug Discov
tumour heterogeneity: a looking glass for can- the formation and maintenance of cancer stem 2006;5:1026–33.
cer? Nat Rev Cancer 2012;12:323–34. cells. Mol Cell 2010;39:761–72. 81. Humke EW, Dorn KV, Milenkovic L, et al. The
43. Biddle A, Liang X, Gammon L, et al. Cancer 61. Shimono Y, Zabala M, Cho RW, et al. Down- output of Hedgehog signaling is controlled by
stem cells in squamous cell carcinoma switch regulation of miRNA-200c links breast cancer the dynamic association between suppressor of

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC
2000 Tumor heterogeneity and CSC paradigm

fused and the Gli proteins. Genes Dev 2010;24: with Hedgehog/GLI in oncogenic transformation 97. Zhang GN, Liang Y, Zhou LJ, et al. Combina-
670–82. via activation of the MEK/ERK/JUN pathway. tion of salinomycin and gemcitabine eliminates
82. Rahman M, Deleyrolle L, Vedam-Mai V, et al. The Cancer Res 2009;69:1284–92. pancreatic cancer cells. Cancer Lett 2011;313:
cancer stem cell hypothesis: failures and pitfalls. 90. Bao S, Wu Q, McLendon RE, et al. Glioma stem 137–44.
Neurosurgery 2011;68:531–45; discussion 45. cells promote radioresistance by preferential 98. Zhang Y, Zhang H, Wang X, et al. The eradica-
83. Molofsky AV, Pardal R, Iwashita T, et al. BMI1 activation of the DNA damage response. Nature tion of breast cancer and cancer stem cells using
dependence distinguishes neural stem cell self- 2006;444:756–60. octreotide modified paclitaxel active targeting
renewal from progenitor proliferation. Nature 91. Ma L, Zhang G, Miao XB, et al. Cancer stem- micelles and salinomycin passive targeting
2003;425:962–7. like cell properties are regulated by EGFR/AKT/ micelles. Biomaterials 2012;33:679–91.
84. Merchant AA, Matsui W. Targeting Hedgehog-a b-catenin signaling and preferentially inhibited 99. Gupta PB, Onder TT, Jiang G, et al. Identifica-
cancer stem cell pathway. Clin Cancer Res 2010; by gefitinib in nasopharyngeal carcinoma. tion of selective inhibitors of cancer stem cells
16:3130–40. FEBS J 2013;280:2027–41. by high-throughput screening. Cell 2009;138:
85. Meng S, Luo M, Sun H, et al. Identification and 92. Tamara Marie-Egyptienne D, Lohse I, Hill RP. 645–59.
characterization of BMI1-responding element Cancer stem cells, the epithelial to mesenchymal 100. Hirsch HA, Iliopoulos D, Struhl K. Metformin
Mini Review

within the human p16 promoter. J Biol Chem transition (EMT) and radioresistance: potential inhibits the inflammatory response associated
2010;285:33219–29. role of hypoxia. Cancer Lett, in press. with cellular transformation and cancer stem
86. Akira S, Nishio Y, Inoue M, et al. Molecular 93. Heddleston JM, Li Z, McLendon RE, et al. The cell growth. Proc Natl Acad Sci USA 2013;110:
cloning of APRF, a novel IFN-stimulated gene hypoxic microenvironment maintains glioblas- 972–7.
factor 3 p91-related transcription factor involved toma stem cells and promotes reprogramming 101. MacKenzie MJ, Ernst S, Johnson C, et al. A
in the gp130-mediated signaling pathway. Cell towards a cancer stem cell phenotype. Cell Cycle phase I study of temsirolimus and metformin in
1994;77:63–71. 2009;8:3274–84. advanced solid tumours. Invest New Drugs 2012;
87. Dimberg LY, Dimberg A, Ivarsson K, et al. Stat1 94. Sugihara E, Saya H. Complexity of cancer stem 30:647–52.
activation attenuates IL-6 induced Stat3 activity cells. Int J Cancer 2013;132:1249–59. 102. Chan CH, Morrow JK, Li CF, et al. Pharmaco-
but does not alter apoptosis sensitivity in multi- 95. Marjanovic ND, Weinberg RA, Chaffer CL. Cell logical inactivation of Skp2 SCF ubiquitin ligase
ple myeloma. BMC Cancer 2012;12:318. plasticity and heterogeneity in cancer. Clin restricts cancer stem cell traits and cancer pro-
88. Vogt PK, Hart JR. PI3K and STAT3: a new alli- Chem 2013;59:168–79. gression. Cell 2013;154:556–68.
ance. Cancer Discov 2011;1:481–6. 96. Naujokat C, Fuchs D, Opelz G. Salinomycin in 103. Harrison C. Anticancer drugs: breaking a can-
89. Schnidar H, Eberl M, Klingler S, et al. Epider- cancer: a new mission for an old agent. Mol cerous interaction. Nat Rev Drug Discov 2013;
mal growth factor receptor signaling synergizes Med Rep 2010;3:555–9. 12:740–1.

Int. J. Cancer: 136, 1991–2000 (2015) V


C 2014 UICC

You might also like