You are on page 1of 10

REvIEWS

Targeting cancer stem cells by inhibiting Wnt,


notch, and Hedgehog pathways
Naoko Takebe, Pamela J. Harris, Ronald Q. Warren and S. Percy Ivy
abstract | Tumor relapse and metastasis remain major obstacles for improving overall cancer survival,
which may be due at least in part to the existence of cancer stem cells (CSCs). CSCs are characterized by
tumorigenic properties and the ability to self-renew, form differentiated progeny, and develop resistance to
therapy. CSCs use many of the same signaling pathways that are found in normal stem cells, such as Wnt,
Notch, and Hedgehog (Hh). The origin of CSCs is not fully understood, but data suggest that they originate
from normal stem or progenitor cells, or possibly other cancer cells. Therapeutic targeting of both CSCs and
bulk tumor populations may provide a strategy to suppress tumor regrowth. Development of agents that
target critical steps in the Wnt, Notch, and Hh pathways will be complicated by signaling cross-talk. The role
that embryonic signaling pathways play in the function of CSCs, the development of new anti-CSC therapeutic
agents, and the complexity of potential CSC signaling cross-talk are described in this Review.
Takebe, N. et al. Nat. Rev. Clin. Oncol. 8, 97–106 (2011); published online 14 December 2010; doi:10.1038/nrclinonc.2010.196

Introduction
tumors are composed of a heterogeneous group of cells, mice.5 CsCs use a variety of signaling pathways to undergo
demonstrated by the fact that some tumor cell fractions self renewal and differentiation, including wnt, notch,
can support new growth in xenograft models, whereas and Hedgehog (Hh).6–8 the slow growth rate and chemo­
other cell fractions do not.1 traditionally, two models have resistant characteristics suggest that CsCs may survive
been proposed to explain tumor cell heterogeneity: the routine chemotherapy, only to reinitiate tumor growth at
stochastic model and the hierarchy model. the stochas­ a later point in time.5
tic model states that tumors arise as a biologically homo­ Given the appropriate combination of Dna mutations
geneous group of cells, with functional heterogeneity and environmental factors, a subpopulation of CsCs
arising through random (that is, stochastic) events.2 in this might acquire metastatic properties. migratory CsCs may
model, tumor initiation may occur in any cell as a result become lodged in distant anatomical sites and continue
of an accumulation of Dna mutations, epigenetic regula­ through self­renewal and asymmetric cell division, to
tion, and a permissive microenvironment.3 the stochastic produce progenitor cells and bulk tumor cells.9 Certain
model suggests that all tumor cells have the potential to tumors, such as melanoma, exhibit high levels of CsCs
become cancer stem cells (CsCs), given the appropriate suggesting that the hierarchical model may not apply
conditions. alternatively, the CsC (or hierarchy) model for all tumors.10 in these cases, additional studies will
suggests that tumors are composed of a heterogeneous be required to determine frequencies of CsCs, perhaps
group of cells that have arisen from stem­like precursors. using assays such as serial dilution experiments in
as these tumor cells differentiate, they form a mixture combination with gene insertion clonality assays, similar
of cells with different biological and phenotypic charac­ to that proposed by von Kalle’s group.11 in brief, CsC
teristics, forming a cellular hierarchy. at the apex of this clonal analysis, using the linear amplification­mediated National Cancer
Institute, Division of
hierarchy are CsCs, which serve as the source of newly PCr (lam­PCr) labeling technique and serial xeno­ Cancer Treatment and
formed tumor cells. graft transplantation, can identify true stem­cells with Diagnosis, Cancer
Therapy Evaluation
a third model, which blends characteristics of the self­renewal properties.12 Program, Investigational
stochastic and CsC models, may provide a mechanism Drug Branch, EPN7131,
for the formation of both primary and metastatic tumors.4 Targeting embryonic signaling pathways 6130 Executive
Boulevard, Rockville,
Chromosomal instability within the CsC population, the first experimental evidence of CsCs came from Bethesda, MD 20852,
together with extrinsic environmental factors, may lead lapidot et al.13 in the 1990s who identified leukemia stem­ USA (n. Takebe,
P. J. Harris, S. P. ivy).
to the appearance of CsC heterogeneity. self­renewing cells capable of initiating human acute myeloid leukemia PSI International, Inc.,
CsCs, comprising a small minority of tumor cells, initi­ (aml) after transplantation into sCiD mice. the investi­ 6500 Rock Spring
ated tumor growth and formed new progenitor and bulk gators further revealed CsCs capable of self­renewal and Drive, Suite 650,
Bethesda, MD 20817,
tumor cells in severe combined immunodeficient (sCiD) production of cancer progenitor cells using a limiting USA (R. Q. Warren).
dilution analysis.14 this in vivo xenograft transplantation
Correspondence to:
competing interests approach to identify cells with self­renewal capability has S. P. Ivy
The authors declare no competing interests. subsequently been applied to identify CsCs in many solid ivyp@ctep.nci.nih.gov

nature reviews | clinical oncology volume 8 | FeBruarY 2011 | 97


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

Key points tumors. in 2004, Clarke and colleagues identified a subset


of rare breast CsCs using a limiting dilution assay in non­
■ The stochastic model and cancer stem cell (CSC) model of tumorigenesis could
obese diabetic sCiD mice that were enriched in CD44+/
be combined to help explain tumor relapse and metastasis
CD24–/lin– cells.15 thereafter, in vivo xenograft limiting
■ DNA mutations, microenvironmental factors, and/or epithelial-to-mesenchymal
dilution analyses were employed to demonstrate CsCs
transition may drive CSCs towards a metastatic phenotype
with tumorigenic activity in a variety of malignancies
■ Tumors composed of small populations of CSCs plus large numbers of bulk
including brain,16 colon,17–19 head and neck,20 pancreas,21,22
tumor cells may be particularly susceptible to combination drug regimens that
target each cell population
hepatocellular,23 melanoma,24 liver,25 lung,26 ovarian,27
prostate,28 bladder,29 and ewing sarcoma.30
■ The potential for cross-talk among signaling pathways by CSCs opens new
opportunities for designing combination drug regimens
owing to the very similar phenotypes of normal stem­
cells and CsCs, efforts to identify and isolate CsCs remain
■ New experimental agents are being developed to block Wnt, Notch, and Hedgehog
signaling by CSCs, some of which are being tested in early clinical trials
a challenge for drug development and treatment strat­
egies. in solid tumors, the identification and enrichment
■ Measurement of biologic effects of anti-CSC therapeutic regimens will remain
of CsCs is dependent on the presence of biomarkers (that
a challenge until more-effective methods to identify CSCs in vivo or in vitro
surrogate assays are improved is, CD133+, CD44+, CD24low, and CD166+). alternatively,
some CsCs can be identified using flow cytometry­based
functional assays such as aldehyde dehydrogenase­1
■ Negative regulatory region mAbs (alDH1) expression and side population assays to enrich
Neighboring
signal-sending cell ■ DLL4 mAbs Ligand degradation
■ Other ligand mAbs or recycling
the CsC population from patient tumor explants.16,31,32
Notch soluble
experimental models suggest that CsCs may originate
Active receptor decoys from normal stem­cells or normal progenitor cells.33,34
ligand several leukemia mouse models have supported this
hypothesis and examples from chronic myeloid leu­
kemia (Cml) clinical samples have indicated that Cml
Delta (DLL1, DLL3, DLL4) progenitor cells could acquire self­renewal capabilities.35
or Jagged (JAG1, JAG2)
nonetheless, tumors often develop and progress due
■ γ-Secretase inhibitors
Extracellular space ■ γ-Secretase modulators to deregulated self­renewal pathways.34 thus, targeting
ADAM/ Nicastrin
these deregulated embryonic signaling pathways holds
Notch TACE Presenilin promise in clinical therapeutic development.
APH-1
PEN-2
notch signaling pathway
Activated Transmembrane γ-Secretase notch signaling has a critical role in regulating cell­to­
portion of Notch complex
receptor cell communication during embryogenesis, cellular
proliferation, differentiation, and apoptosis. 36 notch
Signal-receiving cell S2 cleavage signaling is also critical for normal hematopoiesis, breast
NICD
development, colorectal epithelial maturation, immune
S3 cleavage regulation, and neural stem cell survival.37,38 mammalian­
membrane­bound notch ligands consist of two structur­
Small interfering RNA,
micro RNA to target mRNAs ally distinct families: Delta­like ligands (Dlls) 1, 3 and 4,
and Jagged ligands 1 and 2 that interact with four trans­
Cytoplasm
MAML1 membrane notch receptors (notch 1–4, Figure 1). the
inhibitor pairing of notch ligand–receptors results in coordinated
Notch target genes communication between adjacent cells. the extracellular
HAT ■ HES family region of the notch receptor contains numerous epi­
MAML1 ■ Myc
■ p21 dermal growth factor­like domains that mediate inter­
CSL SKIP
actions with notch ligands. affinity between the notch
Nucleus ligand and receptor depends on the extent of epidermal
growth factor domain fucosylation by the Fringe proteins,
On/derepressed
that is, lunatic, radical, or manic.39 notch receptors exist
Figure 1 | Notch signaling pathway inhibition. Activation of the Notch receptor as hetero dimers, consisting of noncovalently bound
occurs following binding of membrane-bound Delta or Jagged ligands during cell-to- extracellular and transmembrane domains.
cell contact. Following absorption and proteolysis of the heterodimer Notch once ligand–receptor binding occurs, the notch
receptor (by ADAM and γ-secretase complex), a soluble fragment—the NICD—is receptor undergoes a conformational change to expose
released into the cytoplasm. The NICD translocates to the nucleus where it serves a previously protected site to proteolytic cleavage by
as a transcriptional activator of Notch-associated target genes, including HES, Myc metalloprotease and γ­secretase, releasing an extracellular
and p21. Potential therapeutic inhibitors of Notch signaling target events such as
and intracellular fragment, respectively.40 these catalytic
γ-secretase complex proteolysis and transcriptional activation. Abbreviations:
ADAM, A disintegrin and metalloproteinase; CSL, CBF1/Su(H)/Lag-1; DLL, delta-
steps cleave the intracellular­membrane domain and
like ligand; HAT, histone acetyltransferase; MAML1, Mastermind-like 1; mAbs, release the active notch intracellular domain (niCD) into
monoclonal antibodies; NICD, Notch intracellular domain; SKIP, ski-interacting the cytoplasm. niCD undergoes nuclear translocation
protein; TACE, TNF-α-converting enzyme. and binding to the transcription initiation complex

98 | FEBRUARY 2011 | volUmE 8 www.nature.com/nrclinonc


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

and core binding factor­1 (CBF­1), thus modulating Table 1 | Experimental Notch inhibitors
notch­specific gene expression.
agents in development Target Mechanism of action

Investigational Notch pathway-targeting agents γ-Secretase inhibitors: 120,121 Notch homologs Inhibition of Notch
MK0752, RO4929097, Notch ligands cleavage by
inhibition of γ­secretase­mediated notch cleavage is PF–03084,014, LY450139, Other γ-secretase substrates γ-secretase
a primary focus for the development of targeted thera­ BMS-708163
peutics. several pharmaceutical companies have devel­ γ-Secretase modifiers:122 Substrates of γ-secretase Inhibition of Notch
oped γ­secretase inhibitors (Gsis) that are in the early MPC-7869 cleavage by
clinical development. two of these agents (ro4929097 γ-secretase
[roche] and mK0752 [merck]) are in phase i testing after MAML1 inhibitors:48,49 Notch homologs Interference with
evaluating a variety of schedules to determine safe and MAML–CSL–Notch, Other nuclear transcription Notch nuclear
tolerable administration regimens (table 1). the initial antennapedia/dominant–MAML factors that target MAML1 co-activator MAML1

testing of mK0752 in t­cell acute lymphoblastic leukemia Negative regulatory region Individual Notch receptors Interference with
was disappointing due to the Gsi­associated toxicity of gut monoclonal antibodies123 and other Notch ligands ligand-induced Notch
subunit separation
goblet cell hyperplasia and associated secretory diarrhea
that was dose limiting. in a mouse model of t­cell acute DLL4 monoclonal Specific for DLL4 Interference with
antibodies:45,124,125 ligand–receptor
lymphoblastic leukemia, the co­administration of gluco­ OMP–21M18, DLL4 antibody, interaction
corticoids and Gsi resulted in the successful reduction of DLL4
toxic effects in the gut, without impairing efficacy.41 Notch soluble receptor Relatively specific for Notch Interference with
strong evidence supports Gsi treatment of estrogen decoys126,127 homologs ligand–receptor
receptor­positive breast cancer in which estrogen block­ Potential pan-Notch inhibition interaction
ade by anti­estrogens or aromatase inhibitors leads to Abbreviations: DLL, Delta-like ligand; MAML, Mastermind-like.
dependence on notch signaling. rizzo and colleagues
found that estrogen inhibited notch activity was mediated
in part through inhibition of γ­secretase activity.42 the embryonic development. 51 Hyperactivation of this
study suggested that inhibition of notch signaling may be pathway, by either mutation or deregulation, has recently
a useful strategy in breast cancer. single­agent Gsi therapy been recognized to cause tumorigenesis in a wide variety
may also have activity in triple­negative breast cancer, as of tissues. most notably, the discovery of mutations
this tumor type harbors CsC­like characteristics. within the human homolog of the Drosophila patched
Dll4 is a notch ligand involved in the process of angio­ gene (Ptch1) in a rare hereditary form of basal cell carci­
genesis. DLL4 mutations in mice result in severe disrup­ noma (BCC), was the first clue to the involvement of
tion of the vasculature,43 whereas heterozygous deletion this pathway in patients with Gorlin syndrome.52,53 Hh
of DLL4 is in general lethal.44 anti­Dll4 agents are in pathway involvement has been observed in other types of
phase i clinical development. no recommended phase ii non­mutation driven, paracrine deregulation of signal­
doses or schedules have been published. reported com­ ing.54 the most interesting, and still evolving concept,
plications of continuous antibody­mediated inhibition involves the association of Hh signaling with CsCs.
of notch include the development of angiomas and neo­ Hh is released from the cell through a dedicated
vascular tuft formation.45,46 these effects may, however, transmembrane transporter Dispatched after acyla­
be antibody­specific. tion of Hh n­terminus by the enzyme rasp located
agents that target mastermind­like (maml)–Csl– in the endoplasmic reticulum.55 Binding of Hh to the
notch complex formation, which is part of the notch transmembrane receptor Ptch1 initiates signaling via
transcriptional complex, remain in very early preclinical the Hh pathway (Figure 2). Ptch1 inhibits the receptor
development. However, this approach to inhibit notch smoothened (smo) by preventing its localization to the
transcription is unique as stapled α­helical peptides have primary cilium, a nonmotile projection present on most
been used to target this complex.47,48 another interesting vertebrate cells. in the presence of Hh, the Hh–Ptch1
approach is to use a genetically engineered fusion protein complex is internalized, allowing smo activation.
of the Drosophila transcription factor antennapedia with localization of smo to the primary cilium, instead of
the truncated version of maml (antP/Dn maml) the plasma membrane, initiates a signaling cascade in
that behaves in a dominant–negative fashion and inhib­ mammals, leading to the activation of the Gli family of
its notch activation.49 the fusion protein is internalized zinc­finger transcription factors. in vertebrates, there
and transported to the nucleus.49 selective inhibition of are three Gli proteins: Gli1 serves to activate Hh target
notch receptors reduce intestinal toxic effects in rodent genes, Gli2 acts both as an activator and repressor, and
xenograft models and may hold promise in future clini­ Gli3 acts as a repressor of target­gene transcription. Hh
cal development.50 this strategy is appealing as it may signaling seems to be dependent on the relative balance
circumvent some of the off­target adverse events caused of Gli activator and repressor forms.56 in breast CsCs,
by notch inhibition, such as diarrhea. Bmi­1, a transcriptional repressor of polycomb group
of transcription factors, and known to be a key regu­
Hedgehog signaling pathway lator of the self­renewal of normal and leukemic stem
the Hh signaling pathway controls tissue polarity, pat­ cells57—the downstream target in the sonic Hh signaling
terning maintenance, and stem­cell maintenance during pathway—is activated.58 Hh signaling may also have a

nature reviews | clinical oncology volume 8 | FeBruarY 2011 | 99


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

Endoplasmic
were sometimes needed to inhibit cell proliferation,
reticulum and golgi indicating the potential for nonspecific effects.
Hh
apparatus
Skn robotnikinin, a small molecule that binds the extra­
cellular sHh protein, has been isolated from small­
Hh-secreting cell
molecule microarray­based screens.64 targeting sHh
HhN ligands may be an interesting approach for prevent­
Dispatched ing tumor relapse and metastasis. tumor­derived sHh
ligands directly activate signaling in stromal cells and
the specificity of sHh targeting may circumvent the
HhN indian Hh inhibition caused by smo inhibitors, which
Ligands potentially causes skeletal bone deformities in young
(IHh, DHh, SHh) Smo antagonist children. identification of small synthetic molecules
Robotnikinin Hh Protein inhibitors (HPis) 1–4 are of interest. HPi­1
Activated HPI-4
Smo inhibits Gli1/2 activation, HPi­2 and HPi­3 inhibits Gli2
activation, and HPi­4 inhibits formation of cilia, when
HhN
smo is activated, resulting in Gli transcription factor
COS activation.65 emerging preclinical models have demon­
Ptch SuFu strated that Hh signaling can modulate the architecture
Gli1/2/3
of the stromal microenvironment and the smo inhibitor
CDO and brother of CDO β-Arrestin iPi­926 (infinity Pharmaceuticals) can lead to improved
HPI-2/3
Kif3A access of chemotherapeutic agents.66
Gli1/2 Active
HPI 1
Clinical development of Hh pathway
administration of GDC­0449 (Genentech), a small­
Cyclin D, Cyclin E, Myc molecule smo inhibitor, resulted in no dose­limiting
Cytoplasm
Gli1, Ptch, HIP toxicities (Dlts) in patients with advanced solid tumors.
Gli1/2 Active Common adverse events included dysgeusia, hair
loss, nausea, vomiting, anorexia, dyspepsia, weight loss,
Nucleus hyponatremia, and fatigue.67 the GDC­0449 study in
33 patients with advanced BCC reported two complete
Figure 2 | Hedgehog signaling pathway inhibition. In the inactive state, the absence responses and 16 partial responses.68 in addition, there
of Hh leads to inhibition of Smo by the transmembrane receptor Ptch while Gli1/2 are currently three company­sponsored phase ii trials
are phosphorylated and removed from the cytoplasm through proteosomal evaluating the efficacy of GDC­0449 in patients with
degradation. In the active state, Hh is secreted by an adjacent cell and binds to ovarian cancer in remission, advanced colorectal cancer,
Ptch, allowing Smo activation. Gli1/2 are released from the Smo protein complex and advanced BCC.
and translocate to the nucleus, leading to transcriptional activation of Hh-
Four phase i and 10 phase ii clinical trials of GDC­0449
associated genes. New therapeutic agents have been developed that target Hh and
Smo activation and downstream proteins, such as Gli. Abbreviations: COS, costal;
are also being sponsored by the national Cancer institute
Hh, Hedgehog; HIP, Hedgehog interacting protein; HPI, Hedgehog protein inhibitor; and the Cancer therapy evaluation Program. these
Ptch, Patched; Smo, Smoothened; SuFu, suppressor of fused. trials include pediatric and adult patients with recurrent
medulloblastoma, advanced pancreatic tumor (in combi­
nation with gemcitabine), glioblastoma multiforme,
key role in maintenance of CsCs in Cml.59 inhibition of gastric carcinoma (in combination with FolFoX [folinic
Hh signaling by the inhibitor cyclopamine was shown acid, fluorouracil and oxaliplatin]), prostate carcinoma,
to inhibit epithelial­to­mesenchymal transistion (emt) soft tissue sarcoma, breast cancer, chondrosarcoma,
and metastases in pancreatic cancer cell lines.60 multiple myeloma (in post­transplant patients), and
small­cell lung cancer (in combination with cisplatin
Preclinical agents targeting the Hh pathway and etoposide). a trial investigating a novel combina­
Cyclopamine (11­deoxojervine), the prototype of Hh tion therapy to inhibit both Hh and notch signaling
pathway­specific inhibitors, is a plant­derived steroidal pathways in advanced breast cancer and soft­tissue sar­
alkaloid that binds to and deactivates smo.61 a number comas is underway. Bristol–myers squibb, in collabora­
of preclinical studies have been performed using this tion with exelixis, has initiated two phase i clinical trials
agent; however, cells without the smo receptor underwent of a small­molecule smo inhibitor (Bms­833923, Xl139)
apoptosis, therefore caution is needed when interpreting in patients with advanced or metastatic solid tumors.
the results of possible ‘off­target’ effects. other synthetic this agent has been also tested in multiple myeloma in
small molecules that are more potent inhibitors of smo combination with lenalidomide, dexamethasone, and
have been described.62,63 Furthermore, several natural bortezommib, in small­cell lung cancer in combination
and synthetic small­molecule inhibitors of smo are being with carboplatin and etoposide, and in metastatic gastric,
investigated preclinically for antitumor activity (table 2). gastroesophageal, and esophageal adenocarcinoma in
However, these in vitro studies should be interpreted with combination with cisplatin and capecitabine. Clinical
caution because high concentrations of Hh antagonist data from these trials are not yet available.

100 | FEBRUARY 2011 | volUmE 8 www.nature.com/nrclinonc


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

the safety of GDC­0449 was reported in a phase i clin­ Table 2 | Experimental Hedgehog inhibitors
ical trial of patients with metastatic or locally advanced
agents in development Target Mechanism of action
BCC. 68 of 33 evaluable patients, eight instances of
Cyclopamine61 (natural compound) Smo Antagonist
grade 3 adverse events were considered possibly attribu­
table to GDC­0449. these adverse events included Synthetic small molecules
fatigue, hyponatremia, muscle spasm, and atrial fibrilla­ IPI–92666 Smo Antagonist
tion. no Dtls were observed and with the exception of GDC-0449128 (Cur-61414) Smo Antagonist
one grade 3 lymphopenia, andno hematologic adverse
BMS–833923 129
Smo Antagonist
events were observed.
Robotnikinin64 Extracellular sHh Inhibitor of sHh
other Hh pathway inhibitors are also in development.
For example, iPi­926 (infinity Pharmaceuticals), a cyclo­ PF–04449913 Smo Antagonist
pamine­derived inhibitor of the Hh pathway, is being LDE225 Smo Antagonist
evaluated in clinical trials for advanced­stage solid tumors HPI 1–465 Gli1/2 (HPI-1) Antagonist
and metastatic pancreatic cancer, and in a randomized, Gli2 (HPI-2 and HPI-3)
double­blind, placebo­controlled phase ii study with gem­ Cilia (HPI-4)
citabine. PF­04449913 (Pfizer), a smo inhibitor, is cur­ Abbreviations: Hh, Hedgehog; HPI, Hedgehog protein inhibitor; sHh, Sonic hedgehog; Smo, Smoothened.

rently being examined in a phase i study as a single agent


or in combination with dasatinib in patients with Cml. a
new smo antagonist lDe225 (novartis), is being investi­ cancer, and wilms tumor.8,58,77–80 in addition, deregulated
gated in a phase i trial in patients with advanced solid wnt signaling has been associated with CsC activity;
tumors, phase i study in BCC, and in a phase i pediatric specifically, cutaneous CsCs require β­catenin signaling
dose­finding study. to maintain their tumorigenic phenotype.81
a microarray analysis enriched from patients with
Wnt signaling pathway aml demonstrated that the wnt signaling pathway is
wnt proteins consist of 19 highly conserved glycoproteins deregulated in leukemic stem cells when compared with
that serve as ligands for the Frizzled (Fz) transmembrane normal hematopoietic stem cells.82 in leukemic stem cells,
receptor.69 During embryogenesis, wnt proteins direct upregulation of genes encoding the wnt pathway pro­
cell fate determination at various stages of development teins axin and adenomatous polyposis coli are observed
and their signaling acts to regulate the development of a frequently and deregulation of wnt signaling in aml
variety of organ systems including cardiovascular, central stem cells may contribute to leukemogenesis.83
nervous system, renal, and lung.70 in adults, wnt signal­
ing has a key role in the regulation of tissue self­renewal, Preclinical agents targeting the Wnt pathway
particularly in intestinal crypts, hair follicles, and bone a number of experimental agents are currently being
growth plates.71,72 loss­of­function and gain­of­function evaluated for their ability to inhibit wnt signaling
studies in mice with β­catenin (CTNNB1) mutations (table 3). Chen et al.84 identified two new classes of
have demonstrated the importance of the wnt signaling small molecules that inhibit wnt signaling. the first is a
pathway in multiple organ systems.70 membrane­bound acyltransferase that inhibits the acti­
wnt binding to the Fz receptors initiates two distinct vity of Porcupine, a molecule essential for wnt synthesis.
signaling cascades, termed canonical or non­canonical. the second inhibits the destruction of axin, a suppres­
the canonical pathway leads to the accumulation of sor of wnt signaling activity. in addition, the small mol­
β­catenin in the nucleus and subsequent transcriptional ecule iCG­001 (institute for Chemical Genomics), was
activation of targeted genes (Figure 3), but this does not reported to selectively inhibit wnt/β­catenin signaling
occur in the non­canonical pathway. to be functional, by interrupting β­catenin binding to the transcriptional
wnts are lipid modified through palmitoylation. 73 cofactor cyclic amP response element­binding protein
secretion of wnt is carried out by the transmembrane (CBP).85 iCG­001 treatment of colon carcinoma cell lines
protein wntless.74 multiple wnt homologs have demon­ resulted in apoptosis, while sparing normal colonic epi­
strated high­affinity binding to the conserved cysteine­ thelial cells. although iCG­001 specifically blocked the
rich domain of Fz.75 interaction of the wnt proteins with interaction between β­catenin and CBP, no interference
10 known mammalian Fz receptors76 suggests that the with β­catenin­induced p300 signaling was observed,
potential for multiple wnt/Fz pairings may fine tune despite the high degree of sequence homology between
the cellular response to wnt. translocation of β­catenin CBP and p300 (63%).85 iCG­001 seems to initiate the key
from the cytoplasm to the nucleus results in interaction switch from β­catenin and CBP intereaction to β­catenin
with members of the t­cell factor–lymphocyte enhancer and p300 interaction that controls a fundamental stem­
factor family of transcriptional factors and subsequent cell and progenitor cell switching, resulting in cell dif­
activation of wnt targeted genes. ferentiation. 86 several compounds are being tested
preclinically to target the PDZ domain of Disheveled,
Deregulated Wnt signaling in cancer or CSCs nsC668036 and FJ9.87,88 Disheveled is a key protein in
aberrant wnt signaling has been reported in tumors from the wnt signaling pathway that links extracellular signals
patients with hepatocellular carcinoma, hepatoblastoma, and downstream signals and may be able to inhibit both
colorectal cancer, aml, Cml, multiple myeloma, gastric non­canonical and canonical wnt signaling pathways.

nature reviews | clinical oncology volume 8 | FeBruarY 2011 | 101


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

e­cadherin, a membrane­bound glycoprotein involved in


Wnt mAbs Small molecule
inhibitors the adherence of adjacent cells. the loss of e­cadherin in
primary tumor tissue has been linked with tumor meta­
WIF-1 sFRP
stasis and poor prognosis.93,94 wnt, notch and Hh are all
DKKs Wnt Fz receptor known inducers of emt along with niche factors, such as
antibody
LRP inhibitors Fz members of the transforming growth factor­β (tGF­β)
family of cytokines.95–98 interactions between tGF­β and
LRP
the embryonic stem­cell signaling pathways can also
Dvl NSC 668036
have an important role in maintaining the stem­cell­like
characteristics of emt­induced tumor cells.99 thus, tar­
CK1 P Naked
geting embryonic signaling pathways may also inhibit
Dvl-P Dvl emt and metastasis.
Axin stabilizing agents
PP2A
Axin Multiprotein
Chemoresistance of cancer stem cells
APC GSK3 PAR-1
destruction the high frequency of tumor relapse following therapy
β-Catenin
complex suggests the presence of residual CsCs that are resistant
β-Catenin
degradation to conventional therapy.1 an increase in chemoresistant
β-Catenin
CD44+/CD24–/low cells has been demonstrated in Her2­
positive breast cancer following treatment with neo­
Cytoplasm adjuvant chemotherapy.100 in this study, post­treatment
β-Catenin Target genes biopsies were enriched in cells with tumorigenic poten­
■ Myc tial, as demonstrated by their increased potential for
Nucleus ■ Cyclin D1
■ TCF-1 Migration
mammosphere formation in vitro and increased capacity
ICG-001
BCL9 Pygo
NSAIDs ■ PPAR-δ to form tumors in a xenograft model.
■ MMP-7 Adhesion
p300 β-Catenin PNU-74654 ■ Axin-2
several mechanisms can be involved in CsC chemo­
β-Catenin
CBP
■ CD44 resistance and radioresistance, including expression of
Tcf/Lef Tcf/Lef
■ Cox2, etc. the atP­binding cassette superfamily of active drug
transporters.101 members of this transmembrane super­
Promotes cell differentiation Promotes cell proliferation family act as unidirectional cellular pumps and have been
linked with multidrug resistance.102 resistance to anti­
Figure 3 | Wnt/β-catenin signaling pathway inhibition. In the absence of Wnt cancer drugs occurs as a result of reduced levels of drug
signaling, cytoplasmic levels of β-catenin are tightly regulated by a multiprotein accumulation within the CsCs. also, resistance to radia­
destruction complex. β-Catenin levels are kept low through phosphorylation, which tion therapy can be caused by increased expression of
leads to ubiquitinylation and subsequent proteosomal degradation. Binding of Wnt
free­radical scavengers by CsCs. these molecules reduce
to the Fz receptors and LRP co-receptors allows β-catenin to be released from the
intracellular levels of reactive oxygen species following
multiprotein destruction complex. The free β-catenin is translocated to the nucleus
where it acts together with either p300 or CBP as a transcriptional activator of radiation therapy.103 short­lived reactive oxygen species
Wnt-associated genes. Agents that inhibit Wnt/Fz binding and downstream events can cause damage both to Dna and proteins, leading to
are in development. Abbreviations: CBP, cyclic AMP response element-binding cell death. By reducing levels of reactive oxygen species,
protein; Fz, frizzled; LRP, low-density lipoprotein receptor-related protein; mAbs, resistant cells can evade the accumulation of cytotoxic
monoclonal antibodies. effects of radiation therapy.103

Cross-talk among signaling pathways


monoclonal antibodies against wnts, Fz receptor, or instances of cross­talk between the embryonic signaling
secreted Fz­related protein have been tested to inhibit pathways notch, wnt, or Hh and other signaling path­
wnt signaling at the extracellular level, with promising ways have been reported in a variety of cell types.104–106
antitumor activity.89,90 although aberrant activation of an individual pathway
may result in tissue specific carcinogenesis, these path­
Epithelial-to-mesenchymal transition ways rarely operate in isolation. Cross­talk between
emt originally defined a process of cellular reorganiza­ signaling pathways has the potential to profoundly add
tion essential for embryonic development. this multi­step to the complexity of cellular responses to external stimuli.
process results in the loss of cell­to­cell adhesive prop­ a link between murine mammalian target of rapamycin
erties, loss of cell polarity, and the gain of invasive and (mtor) and notch signaling pathways shows that cells
migratory mesenchymal properties.91 During embryo­ with constitutively activated mtor also express ele­
genesis, emt allows progenitor cells to migrate to distant vated levels of niCD and Hes1, suggesting that notch
sites within an embryo to form new tissue.92 the emt is upregulated by mtor.107 treatment with the mtor
process is reversible, with mesenchymal­to­epithelial inhibitor rapamycin led to decreased signaling of both
transition allowing cells with mesenchymal characteris­ mtor and notch. upregulation of notch by mtor acts
tics to revert to epithelial­like cells. the emt processes through the stat3/p63/Jagged pathway. Conversely,
also occur during tumorigenesis, allowing some CsCs downregulation of notch signaling inhibited mtor, akt,
to become metastatic. During emt, cells downregulate and nuclear factor­κB signaling.108 Cross­talk between

102 | FEBRUARY 2011 | volUmE 8 www.nature.com/nrclinonc


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

notch and the Pi3K/akt pathways was reported in a Table 3 | Experimental Wnt inhibitors
variety of cell types.38,109
agents in development Target Mechanism of action
interactions between eGFr and notch signaling have
been reported in breast cancer cell lines.110 treatment Wnt monoclonal antibodies Wnt ligands Ligand binding89,90
Soluble Wnt receptor (Fz 8
of these cells in vitro with gefitinib, an eGFr inhibi­ cysteine-rich domain fused to the
tor, resulted in decreased expression of both eGFr and human Fc domain) antibody
notch­1. notch­1 knockdown with small interfering (decoy receptor)
rna in glioma cell lines led to decreased eGFr mes­ Fz receptor antibody Fz receptors Inhibition of ligand-receptor
senger rna and protein expression. 111 silencing of interaction130
notch expression was also associated with decreased NSC668036 FJ9 (disrupts Fz 7 Disheveled PDZ Inhibition of disheveled PDZ
expression of p53, a known transcriptional activator and Disheveled) domain domain, causing stabilization of
β-catenin degradation complex87,88
of eGFr.112 eGFr is overexpressed in a large propor­
tion of gliomas;113 thus, combination therapies targeting Thiazolidinedione β-Catenin reverse Transportation of β-catenin from
both eGFr and notch may have clinical benefits for this nuclear transport the nucleus131

patient population. PNU-74654 β-Catenin Inhibition of protein-protein


an inverse correlation has been observed between ICG-001 (cyclic AMP response T-cell factor interactions resulting in
element-binding protein β-catenin) decreased β-catenin-dependent
notch and Her2 signaling. Breast cancer cell lines that NSAIDs (e.g., Cox2 inhibitor) gene expression85,132,133
overexpress Her2 had 6­fold to 20­fold lower levels of
Av65 Protein Mechanism unknown, possibly
notch transcriptional activity than cells expressing low Artificial F-box protein degradation β-catenin proteosomal
levels of Her2, suggesting that Her2 overexpression acts Sulindac process degradation84,134–137
to suppress notch signaling.114 By contrast, trastuzumab­ Inhibitors of Wnt response Axin2 Interacts with Axin2 to stabilize
mediated suppression of Her2 phosphorylation resulted Axin proteins causing β-catenin
in increased notch nuclear accumulation and increased loss84
notch expression. alternative notch signaling may occur Inhibitors of Wnt production Porcupine Inhibition of Wnt secretion84
in trastuzumab­resistant breast cancer cells. an analysis Abbreviation: Fz, Frizzled.
of 48 primary breast tumors revealed a significant link
between notch 1 and 3 expression and Her2­negative
tumors.115 expression of notch in Her2­negative breast Conclusions
tumors may represent a novel target for antitumor strat­ the combined stochastic–CsC model provides a possible
egies. Cross­talk between the notch and Her2 pathways explanation for the initiation of tumorigenesis, tumor
in breast cancer may provide cells with a mechanism for relapse, and metastasis in the majority of tumors. this
trastuzumab resistance. model suggests that CsCs evolve into cells with meta­
reports indicate that cross­talk occurs between static potential through accumulated somatic mutations,
tGF­β and each of the embryonic signaling pathways. emt, or stromal microenvironmental factors. tumors
For example, tGF­β­induced emt could be blocked by composed of small populations of CsCs intermixed with
rna silencing of either the Hey1 or Jag1 genes, suggest­ large numbers of bulk tumor cells may require alterna­
ing that there is cross­talk between tGF­β and notch tive treatment strategies to effectively inhibit growth and
pathways.116 tGF­β­induced emt was observed in epi­ prevent relapse. For this reason, CsCs are increasingly
thelial cells. the cellular process of acquiring a metastatic becoming priority targets for the development of new
phenotype was mediated through the induction of the antitumor therapeutics.
notch ligand Jagged­1 and the notch target gene Hey1 these new approaches to cancer therapy will be accom­
in epithelial cells.117 panied by significant challenges. First among these is the
Cross­talk between the wnt and Hh signaling pathways apparent lack of universal stem­cell­specific markers
can also occur through sFrP­1, expression of which is to identify CsCs from different tissues. Putative CsCs
induced by Gli1 in gastric cancer cell lines.118 Hh signaling were successfully enriched using cell surface phenotype
was found to inhibit wnt signaling through upregulation in some preliminary pioneering work in this area, but
of sFrP­1.118 interaction with Hh signaling and notch more recent data have demonstrated that CsCs undergo
may be important in breast cancer as Hh may modulate dynamic changes at the level of cell­surface markers.119
the tumorigenic property in cooperation with notch. these changes are possibly due to epigenetic regulation
Hh signaling activation induces Jagged 2, a notch ligand and growth factors controlled by the microenvironment,
resulting in upregulation of the notch pathway. this making it difficult to define the CsC subpopulation on
interaction is also potentiated by active eGFr signaling as the basis of cell­surface marker expression. the identifi­
described above. signaling pathways, instead of acting cation of CsC­associated markers and pharmaco­
as isolated units, may interact through cross­talk as part dynamic marker assays to monitor the therapeutic effects
of a broader signaling network. these cross­talk inter­ in patients are critical hurdles. ideally, in vivo imaging
actions may contribute to the cellular diversity associated of CsCs would be of interest to monitor the biological
with stem cells during embryogenesis and tissue main­ effects of experimental agents, but these screens are not
tenance. the identification of specific cross­talk net­ yet available.
works in tumor cells may also allow more effective use of signaling pathways can interact through cross­talk as
combination antitumor therapies. part of broad, complex signaling networks. Cross­talk

nature reviews | clinical oncology volume 8 | FeBruarY 2011 | 103


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

may also contribute to the cellular diversity associated a more­effective way of inhibiting tumor relapse and
with stem cells during embryogenesis and tissue main­ metastasis. targeting of embryonic signaling pathways,
tenance, and may have a key role in the growth of CsCs. used by CsCs, represents an emerging and potentially
the identification of cross­talk networks can provide new beneficial new field of cancer therapeutics.
opportunities for designing more­effective treatment
regimens. new experimental agents are being developed Review criteria
to inhibit the wnt, notch, and Hh signaling pathways, Data for this Review were compiled by searching PubMed,
which are frequently utilized by stem cells. Proof­of­ Google Scholar, and Clinicaltrials.gov databases. The
concept clinical trials are currently underway for many search terms included “Notch”, “Wnt”, “Hedgehog”,
of these agents, including GDC­0449, ro4929097, and “cancer stem cells”, “NOD-SCID”, “embryonic signaling
several Hh and notch pathway inhibitors. although still pathway”, “self-renewal”, “EMT”, “migrating cancer stem
early in development, this new approach of targeting cells”, “γ-secretase inhibitor”, “TGF-β”, and “BMP”. In
general, we selected full text articles published in English.
both the bulk tumor and CsC populations may provide

1. Dick, J. E. Stem cell concepts renew cancer 19. Ricci-vitiani, L. et al. Identification and expansion 34. Reya, T., Morrison, S. J., Clarke, M. F. &
research. Blood 112, 4793–4807 (2008). of human colon-cancer-initiating cells. Nature Weissman, I. L. Stem cells, cancer, and cancer
2. Dick, J. E. Looking ahead in cancer stem cell 445, 111–115 (2007). stem cells. Nature 414, 105–111 (2001).
research. Nat. Biotechnol. 27, 44–46 (2009). 20. Prince, M. E. et al. Identification of a 35. Jamieson, C. H. et al. Granulocyte-macrophage
3. Johnsen, H. E. et al. Cancer stem cells and the subpopulation of cells with cancer stem cell progenitors as candidate leukemic stem cells in
cellular hierarchy in haematological properties in head and neck squamous cell blast-crisis CML. N. Engl. J. Med. 351, 657–667
malignancies. Eur. J. Cancer 45 (Suppl. 1), carcinoma. Proc. Natl Acad. Sci. USA 104, (2004).
194–201 (2009). 973–978 (2007). 36. Artavanis-Tsakonas, S., Rand, M. D. & Lake, R. J.
4. Odoux, C. et al. A stochastic model for cancer 21. Hermann, P. C. et al. Distinct populations of Notch signaling: cell fate control and signal
stem cell origin in metastatic colon cancer. cancer stem cells determine tumor growth and integration in development. Science 284,
Cancer Res. 68, 6932–6941 (2008). metastatic activity in human pancreatic cancer. 770–776 (1999).
5. Frank, N. Y., Schatton, T. & Frank, M. H. The Cell Stem Cell 1, 313–323 (2007). 37. Dontu, G. et al. Role of Notch signaling in cell-
therapeutic promise of the cancer stem cell 22. Li, C. et al. Identification of pancreatic cancer fate determination of human mammary stem/
concept. J. Clin. Invest. 120, 41–50 (2010). stem cells. Cancer Res. 67, 1030–1037 (2007). progenitor cells. Breast Cancer Res. 6,
6. McGovern, M., voutev, R., Maciejowski, J., 23. Yamashita, T. et al. EpCAM-positive R605–R615 (2004).
Corsi, A. K. & Hubbard, E. J. A “latent niche” hepatocellular carcinoma cells are tumor- 38. Androutsellis-Theotokis, A. et al. Notch signalling
mechanism for tumor initiation. Proc. Natl Acad. initiating cells with stem/progenitor cell regulates stem cell numbers in vitro and in vivo.
Sci. USA 106, 11617–11622 (2009). features. Gastroenterology 136, 1012–1024 Nature 442, 823–826 (2006).
7. Peacock, C. D. et al. Hedgehog signaling (2009). 39. Rampal, R., Arboleda-velasquez, J. F.,
maintains a tumor stem cell compartment in 24. Schatton, T. et al. Identification of cells initiating Nita-Lazar, A., Kosik, K. S. & Haltiwanger, R. S.
multiple myeloma. Proc. Natl Acad. Sci. USA 104, human melanomas. Nature 451, 345–349 Highly conserved O-fucose sites have distinct
4048–4053 (2007). (2008). effects on Notch1 function. J. Biol. Chem. 280,
8. Reya, T. & Clevers, H. Wnt signalling in stem 25. Yang, Z. F. et al. Significance of CD90+ cancer 32133–32140 (2005).
cells and cancer. Nature 434, 843–850 (2005). stem cells in human liver cancer. Cancer Cell 13, 40. Gordon, W. R. et al. Structural basis for
9. Brabletz, T., Jung, A., Spaderna, S., Hlubek, F. 153–166 (2008). autoinhibition of Notch. Nat. Struct. Mol. Biol.
& Kirchner, T. Opinion: migrating cancer stem 26. Bertolini, G. et al. Highly tumorigenic lung cancer 14, 295–300 (2007).
cells—an integrated concept of malignant CD133+ cells display stem-like features and are 41. Real, P. J. et al. Gamma-secretase inhibitors
tumour progression. Nat. Rev. Cancer 5, spared by cisplatin treatment. Proc. Natl Acad. reverse glucocorticoid resistance in T cell acute
744–749 (2005). Sci. USA 106, 16281–16286 (2009). lymphoblastic leukemia. Nat. Med. 15, 50–58
10. Quintana, E. et al. Efficient tumour formation 27. Zhang, S. et al. Identification and (2009).
by single human melanoma cells. Nature 456, characterization of ovarian cancer-initiating cells 42. Rizzo, P. et al. Rational targeting of Notch signaling
593–598 (2008). from primary human tumors. Cancer Res. 68, in cancer. Oncogene 27, 5124–5131 (2008).
11. Dieter, S. M. et al. Distinct types of human colon 4311–4320 (2008). 43. Duarte, A. et al. Dosage-sensitive requirement
cancer initiating cells contribute to primary 28. Wang, X. et al. A luminal epithelial stem cell that for mouse Dll4 in artery development. Genes
tumor and metastasis formation [abstract 9]. is a cell of origin for prostate cancer. Nature 461, Dev. 18, 2474–2478 (2004).
Proc. Am. Assoc. Cancer Res. (2010). 495–500 (2009). 44. Suchting, S. et al. The Notch ligand Delta-like 4
12. Schmidt, M. et al. Polyclonal long-term 29. Chan, K. S. et al. Identification, molecular negatively regulates endothelial tip cell
repopulating stem cell clones in a primate characterization, clinical prognosis, and formation and vessel branching. Proc. Natl Acad.
model. Blood 100, 2737–2743 (2002). therapeutic targeting of human bladder tumor- Sci. USA 104, 3225–3230 (2007).
13. Lapidot, T. et al. A cell initiating human acute initiating cells. Proc. Natl Acad. Sci. USA 106, 45. Ridgway, J. et al. Inhibition of Dll4 signalling
myeloid leukaemia after transplantation into 14016–14021 (2009). inhibits tumour growth by deregulating
SCID mice. Nature 367, 645–648 (1994). 30. Suvà, M. L. et al. Identification of cancer stem angiogenesis. Nature 444, 1083–1087 (2006).
14. Bonnet, D. & Dick, J. E. Human acute myeloid cells in Ewing’s sarcoma. Cancer Res. 69, 46. Yan, M. et al. Chronic DLL4 blockade induces
leukemia is organized as a hierarchy that 1776–1781 (2009). vascular neoplasms. Nature 463, E6–E7 (2010).
originates from a primitive hematopoietic cell. 31. Huang, E. H. et al. Aldehyde dehydrogenase 1 47. Moellering, R. E. et al. Direct inhibition of the
Nat. Med. 3, 730–737 (1997). is a marker for normal and malignant human NOTCH transcription factor complex. Nature
15. Al-Hajj, M. et al. Prospective identification of colonic stem cells (SC) and tracks SC 462, 182–188 (2009).
tumorigenic breast cancer cells. Proc. Natl Acad. overpopulation during colon tumorigenesis. 48. Sawyer, T. K. AILERON Therapeutics. Chem. Biol.
Sci. USA 100, 3983–3988 (2003). Cancer Res. 69, 3382–3389 (2009). Drug Des. 73, 3–6 (2009).
16. Singh, S. K. et al. Identification of human brain 32. Wang, Z. et al. Down-regulation of notch-1 49. Epenetos, A., Kousparou, C. & Stylianou, S.
tumour initiating cells. Nature 432, 396–401 inhibits invasion by inactivation of nuclear Inhibition of Notch signaling for the treatment
(2004). factor-kappaB, vascular endothelial growth of human carcinomas. AACR Meeting Abstracts
17. Dalerba, P. et al. Phenotypic characterization of factor, and matrix metalloproteinase-9 in A5502 (2009).
human colorectal cancer stem cells. Proc. Natl pancreatic cancer cells. Cancer Res. 66, 50. Wu, Y. et al. Therapeutic antibody targeting
Acad. Sci. USA 104, 10158–10163 (2007). 2778–2784 (2006). of individual Notch receptors. Nature 464,
18. O’Brien, C. A., Pollett, A., Gallinger, S. & 33. Krivtsov, A. v. et al. Transformation from 1052–1057 (2010).
Dick, J. E. A human colon cancer cell capable of committed progenitor to leukaemia stem cell 51. Ingham, P. W. & McMahon, A. P. Hedgehog
initiating tumour growth in immunodeficient initiated by MLL-AF9. Nature 442, 818–822 signaling in animal development: paradigms and
mice. Nature 445, 106–110 (2007). (2006). principles. Genes Dev. 15, 3059–3087 (2001).

104 | FEBRUARY 2011 | volUmE 8 www.nature.com/nrclinonc


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

52. Hahn, H. et al. Mutations of the human homolog growth plate: regulation with chondrocyte 93. Kim, M. A. et al. Prognostic importance of
of Drosophila patched in the nevoid basal differentiation. Bone 40, 1361–1369 (2007). epithelial-mesenchymal transition-related
cell carcinoma syndrome. Cell 85, 841–851 73. Takada, R. et al. Monounsaturated fatty acid protein expression in gastric carcinoma.
(1996). modification of Wnt protein: its role in Wnt Histopathology 54, 442–451 (2009).
53. Johnson, R. L. et al. Human homolog of patched, secretion. Dev. Cell 11, 791–801 (2006). 94. Mareel, M. et al. E-cadherin/catenin/
a candidate gene for the basal cell nevus 74. Ching, W. & Nusse, R. A dedicated Wnt secretion cytoskeleton complex: a regulator of cancer
syndrome. Science 272, 1668–1671 (1996). factor. Cell 125, 432–433 (2006). invasion. J. Cell Physiol. 173, 271–274 (1997).
54. Yauch, R. L. et al. A paracrine requirement for 75. Hsieh, J. C., Rattner, A., Smallwood, P. M. & 95. Dissanayake, S. K. et al. The Wnt5A/protein
hedgehog signalling in cancer. Nature 455, Nathans, J. Biochemical characterization of Wnt- kinase C pathway mediates motility in melanoma
406–410 (2008). frizzled interactions using a soluble, biologically cells via the inhibition of metastasis
55. Micchelli, C. A., The, I., Selva, E., Mogila, v. & active vertebrate Wnt protein. Proc. Natl Acad. suppressors and initiation of an epithelial to
Perrimon, N. Rasp, a putative transmembrane Sci. USA 96, 3546–3551 (1999). mesenchymal transition. J. Biol. Chem. 282,
acyltransferase, is required for Hedgehog 76. Schulte, G. & Bryja, v. The Frizzled family of 17259–17271 (2007).
signaling. Development 129, 843–851 (2002). unconventional G-protein-coupled receptors. 96. vincan, E. & Barker, N. The upstream
56. Ruiz i Altaba, A., Mas, C. & Stecca, B. The Gli Trends Pharmacol. Sci. 28, 518–525 (2007). components of the Wnt signalling pathway
code: an information nexus regulating cell fate, 77. de La Coste, A. et al. Somatic mutations of the in the dynamic EMT and MET associated with
stemness and cancer. Trends Cell Biol. 17, beta-catenin gene are frequent in mouse and colorectal cancer progression. Clin. Exp.
438–447 (2007). human hepatocellular carcinomas. Proc. Natl Metastasis 25, 657–663 (2008).
57. Molofsky, A. v. et al. Bmi-1 dependence Acad. Sci. USA 95, 8847–8851 (1998). 97. Massagué, J. TGFbeta in cancer. Cell 134,
distinguishes neural stem cell self-renewal from 78. Kim, M. S., Kim, S. S., Ahn, C. H., Yoo, N. J. & 215–230 (2008).
progenitor proliferation. Nature 425, 962–967 Lee, S. H. Frameshift mutations of Wnt pathway 98. Yang, J. & Weinberg, R. A. Epithelial-
(2003). genes AXIN2 and TCF7L2 in gastric carcinomas mesenchymal transition: at the crossroads of
58. Adesina, A. M. et al. Gene expression profiling with high microsatellite instability. Hum. Pathol. development and tumor metastasis. Dev. Cell
reveals signatures characterizing histologic 40, 58–64 (2009). 14, 818–829 (2008).
subtypes of hepatoblastoma and global 79. Koesters, R. et al. Mutational activation of the 99. Bailey, J. M., Singh, P. K. & Hollingsworth, M. A.
deregulation in cell growth and survival beta-catenin proto-oncogene is a common event Cancer metastasis facilitated by developmental
pathways. Hum. Pathol. 40, 843–853 (2009). in the development of Wilms’ tumors. Cancer pathways: Sonic hedgehog, Notch, and bone
59. Zhao, C. et al. Hedgehog signaling is essential Res. 59, 3880–3882 (1999). morphogenic proteins. J. Cell Biochem. 102,
for maintenance of cancer stem cells in myeloid 80. Martin, v. et al. Epigenetic regulation of the 829–839 (2007).
leukaemia. Nature 458, 776–779 (2009). non-canonical Wnt pathway in acute myeloid 100. Li, X. et al. Intrinsic resistance of tumorigenic
60. Feldmann, G. et al. Blockade of hedgehog leukemia. Cancer Sci. 101, 425–432 (2010). breast cancer cells to chemotherapy. J. Natl
signaling inhibits pancreatic cancer invasion and 81. Malanchi, I. et al. Cutaneous cancer stem cell Cancer Inst. 100, 672–679 (2008).
metastases: a new paradigm for combination maintenance is dependent on beta-catenin 101. Frank, N. Y. et al. ABCB5-mediated doxorubicin
therapy in solid cancers. Cancer Res. 67, signalling. Nature 452, 650–653 (2008). transport and chemoresistance in human
2187–2196 (2007). 82. Majeti, R. et al. Dysregulated gene expression malignant melanoma. Cancer Res. 65,
61. Taipale, J. et al. Effects of oncogenic mutations networks in human acute myelogenous leukemia 4320–4333 (2005).
in Smoothened and Patched can be reversed stem cells. Proc. Natl Acad. Sci. USA 106, 102. Ambudkar, S. v., Kimchi-Sarfaty, C., Sauna, Z. E.
by cyclopamine. Nature 406, 1005–1009 3396–3401 (2009). & Gottesman, M. M. P-glycoprotein: from
(2000). 83. Müller-Tidow, C. et al. Translocation products in genomics to mechanism. Oncogene 22,
62. Kiselyov, A. S. Targeting the hedgehog signaling acute myeloid leukemia activate the Wnt 7468–7485 (2003).
pathway with small molecules. Anticancer Agents signaling pathway in hematopoietic cells. Mol. 103. Diehn, M. et al. Association of reactive oxygen
Med. Chem. 6, 445–449 (2006). Cell Biol. 24, 2890–2904 (2004). species levels and radioresistance in cancer
63. Rubin, L. L. & de Sauvage, F. J. Targeting the 84. Chen, B. et al. Small molecule-mediated stem cells. Nature 458, 780–783 (2009).
Hedgehog pathway in cancer. Nat. Rev. Drug disruption of Wnt-dependent signaling in tissue 104. Itasaki, N. & Hoppler, S. Crosstalk between Wnt
Discov. 5, 1026–1033 (2006). regeneration and cancer. Nat. Chem. Biol. 5, and bone morphogenic protein signaling:
64. Stanton, B. Z. et al. A small molecule that binds 100–107 (2009). a turbulent relationship. Dev. Dyn. 239, 16–33
Hedgehog and blocks its signaling in human 85. Emami, K. H. et al. A small molecule inhibitor of (2010).
cells. Nat. Chem. Biol. 5, 154–156 (2009). beta-catenin/CREB-binding protein transcription 105. Sun, L., Tian, Z. & Wang, J. A direct cross-talk
65. Hyman, J. M. et al. Small-molecule inhibitors [corrected]. Proc. Natl Acad. Sci. USA 101, between interferon-gamma and sonic hedgehog
reveal multiple strategies for Hedgehog pathway 12682–12687 (2004). signaling that leads to the proliferation of
blockade. Proc. Natl Acad. Sci. USA 106, 86. Takahashi-Yanaga, F. & Kahn, M. Targeting Wnt neuronal precursor cells. Brain Behav. Immun.
14132–14137 (2009). signaling: can we safely eradicate cancer stem 24, 220–228 (2010).
66. Olive, K. P. et al. Inhibition of Hedgehog signaling cells? Clin. Cancer Res. 16, 3153–3162 106. vivekanand, P. & Rebay, I. Intersection of signal
enhances delivery of chemotherapy in a mouse (2010). transduction pathways and development. Annu.
model of pancreatic cancer. Science 324, 87. Fujii, N. et al. An antagonist of dishevelled Rev. Genet. 40, 139–157 (2006).
1457–1461 (2009). protein-protein interaction suppresses 107. Ma, J. et al. Mammalian target of rapamycin
67. LoRusso, P. M. et al. A first-in-human, beta-catenin-dependent tumor cell growth. regulates murine and human cell differentiation
first-in-class, phase (ph) I study of systemic Cancer Res. 67, 573–579 (2007). through STAT3/p63/Jagged/Notch cascade.
Hedgehog (Hh) pathway antagonist, GDC-0449, 88. Shan, J., Shi, D. L., Wang, J. & Zheng, J. J. Clin. Invest. 120, 103–114 (2010).
in patients (pts) with advanced solid tumors Identification of a specific inhibitor of the 108. Wang, Z. et al. Down-regulation of Notch-1 and
[abstract]. J. Clin. Oncol. 26, a3516 (2008). dishevelled PDZ domain. Biochemistry 44, Jagged-1 inhibits prostate cancer cell growth,
68. von Hoff, D. D. et al. Inhibition of the hedgehog 15495–15503 (2005). migration and invasion, and induces apoptosis
pathway in advanced basal-cell carcinoma. 89. He, B. et al. Blockade of Wnt-1 signaling induces via inactivation of Akt, mTOR, and NF-kappaB
N. Engl. J. Med. 361, 1164–1172 (2009). apoptosis in human colorectal cancer cells signaling pathways. J. Cell. Biochem. 109,
69. Angers, S. & Moon, R. T. Proximal events in Wnt containing downstream mutations. Oncogene 726–736 (2010).
signal transduction. Nat. Rev. Mol. Cell Biol. 10, 24, 3054–3058 (2005). 109. Meurette, O. et al. Notch activation induces Akt
468–477 (2009). 90. You, L. et al. An anti-Wnt-2 monoclonal antibody signaling via an autocrine loop to prevent
70. Grigoryan, T., Wend, P., Klaus, A. & Birchmeier, W. induces apoptosis in malignant melanoma cells apoptosis in breast epithelial cells. Cancer Res.
Deciphering the function of canonical Wnt and inhibits tumor growth. Cancer Res. 64, 69, 5015–5022 (2009).
signals in development and disease: conditional 5385–5389 (2004). 110. Dai, J. et al. Cross-talk between Notch and EGFR
loss- and gain-of-function mutations of beta- 91. Thiery, J. P., Acloque, H., Huang, R. Y. & signaling in human breast cancer cells. Cancer
catenin in mice. Genes Dev. 22, 2308–2341 Nieto, M. A. Epithelial-mesenchymal transitions Invest. 27, 533–540 (2009).
(2008). in development and disease. Cell 139, 871–890 111. Purow, B. W. et al. Notch-1 regulates transcription
71. Clevers, H. Wnt/beta-catenin signaling in (2009). of the epidermal growth factor receptor through
development and disease. Cell 127, 469–480 92. Shook, D. & Keller, R. Mechanisms, mechanics p53. Carcinogenesis 29, 918–925 (2008).
(2006). and function of epithelial-mesenchymal 112. Deb, S. P., Muñoz, R. M., Brown, D. R.,
72. Andrade, A. C., Nilsson, O., Barnes, K. M. & transitions in early development. Mech. Dev. Subler, M. A. & Deb, S. Wild-type human p53
Baron, J. Wnt gene expression in the post-natal 120, 1351–1383 (2003). activates the human epidermal growth factor

nature reviews | clinical oncology volume 8 | FeBruarY 2011 | 105


© 2011 Macmillan Publishers Limited. All rights reserved
RevieWS

receptor promoter. Oncogene 9, 1341–1349 122. Kopan, R. & Ilagan, M. X. Gamma-secretase: gamma ligand, inhibits growth and metastasis of
(1994). proteasome of the membrane? Nat. Rev. Mol. HT-29 human colon cancer cells through
113. Mellinghoff, I. K. et al. Molecular determinants of Cell Biol. 5, 499–504 (2004). differentiation-promoting effects. Int. J. Oncol.
the response of glioblastomas to EGFR kinase 123. Li, K. et al. Modulation of Notch signaling by 25, 631–639 (2004).
inhibitors. N. Engl. J. Med. 353, 2012–2024 antibodies specific for the extracellular negative 132. Arber, N. et al. Celecoxib for the prevention of
(2005). regulatory region of NOTCH3. J. Biol. Chem. 283, colorectal adenomatous polyps. N. Engl. J. Med.
114. Osipo, C. et al. ErbB-2 inhibition activates 8046–8054 (2006). 355, 885–895 (2006).
Notch-1 and sensitizes breast cancer cells to 124. Hoey, T. et al. DLL4 blockade inhibits tumor 133. van Stolk, R. et al. Phase I trial of exisulind
a gamma-secretase inhibitor. Oncogene 27, growth and reduces tumor-initiating cell (sulindac sulfone, FGN-1) as a chemopreventive
5019–5032 (2008). frequency. Cell Stem Cell 5, 168–177 (2009). agent in patients with familial adenomatous
115. Hirose, H. et al. Notch pathway as candidate 125. Noguera-Troise, I. et al. Blockade of Dll4 inhibits polyposis. Clin. Cancer Res. 6, 78–89 (2000).
therapeutic target in Her2/Neu/ErbB2 receptor- tumour growth by promoting non-productive 134. Cong, F., Zhang, J., Pao, W., Zhou, P. &
negative breast tumors. Oncol. Rep. 23, 35–43 angiogenesis. Nature 444, 1032–1037 (2006). varmus, H. A protein knockdown strategy to
(2010). 126. Garcés, C. et al. Notch-1 controls the expression study the function of beta-catenin in
116. Guo, X. & Wang, X. F. Signaling cross-talk between of fatty acid-activated transcription factors and tumorigenesis. BMC Mol. Biol. 4, 10 (2003).
TGF-beta/BMP and other pathways. Cell Res. 19, is required for adipogenesis. J. Biol. Chem. 272, 135. Liu, J., Stevens, J., Matsunami, N. & White, R. L.
71–88 (2009). 29729–29734 (1997). Targeted degradation of beta-catenin by
117. Zavadil, J., Cermak, L., Soto-Nieves, N. & 127. Nickoloff, B. J. et al. Jagged-1 mediated chimeric F-box fusion proteins. Biochem.
Böttinger, E. P. Integration of TGF-beta/Smad activation of notch signaling induces complete Biophys. Res. Commun. 313, 1023–1029
and Jagged1/Notch signalling in epithelial-to- maturation of human keratinocytes through (2004).
mesenchymal transition. EMBO J. 23, NF-kappaB and PPARgamma. Cell Death Differ. 9, 136. Nagao, R. et al. A novel β-catenin inhibitor, Av65
1155–1165 (2004). 842–855 (2002). suppresses the growth of CML cell lines which
118. He, J. et al. Suppressing Wnt signaling by the 128. Rudin, C. M., Hann, C. L., Peacock, C. D. & acquire imatinib-resistance because of Abl
hedgehog pathway through sFRP-1. J. Biol. Chem. Watkins, D. N. Novel systemic therapies for kinase domain mutations including T315I and
281, 35598–35602 (2006). small cell lung cancer. J. Natl Compr. Canc. Netw. hypoxia-adaptation. 50th ASH Annual Meeting
119. Bhagwandin, v. J. & Shay. J. W. Pancreatic cancer 6, 315–322 (2008). and Exposition Abstracts, 1081 (2008).
stem cells: fact or fiction? Biochim. Biophys. Acta 129. Barginear, M. F., Leung, M. & Budman, D. R. The 137. Su, Y., Ishikawa, S., Kojima, M. & Liu, B.
1792, 248–259 (2009). hedgehog pathway as a therapeutic target for Eradication of pathogenic beta-catenin by Skp1/
120. Deangelo, D. J. et al. A phase I clinical trial of the treatment of breast cancer. Breast Cancer Res. Cullin/F box ubiquitination machinery. Proc. Natl
notch inhibitor MK-0752 in patients with T-cell Treat. 116, 239–246 (2009). Acad. Sci. USA 100, 12729–12734 (2003).
acute lymphoblastic leukemia/lymphoma (T-ALL) 130. Fukukawa, C. Katagiri, T., Nakatsuru, S. &
and other leukemias [abstract]. J. Clin. Oncol. 24, Nakamura, Y. Therapeutic potential of antibodies
a6585 (2006). against frizzled homologue 10, a cell-surface author contributions
121. Peters, J. U. et al. Novel orally active, protein, for synovial sarcoma [abstract]. Proc. N. Takebe, P. J. Harris, R. Q. Warren and S. P. Ivy
dibenzazepinone-based gamma-secretase Amer. Assoc. Cancer Res. 47, 1975 (2006). contributed equally to the literature review,
inhibitors. Bioorg Med. Chem. Lett. 17, 131. Yoshizumi, T. et al. Thiazolidinedione, a discussions of the content, writing and reviewing and
5918–5923 (2007). peroxisome proliferator-activated receptor- editing of this manuscript before submission.

106 | FEBRUARY 2011 | volUmE 8 www.nature.com/nrclinonc


© 2011 Macmillan Publishers Limited. All rights reserved

You might also like