Вы находитесь на странице: 1из 14

Journal of Pathology

J Pathol 2011; 223: 205–218 INVITED REVIEW


Published online 18 October 2010 in Wiley Online Library
(wileyonlinelibrary.com) DOI: 10.1002/path.2785

The dynamic roles of TGF-β in cancer


Erik Meulmeester1 and Peter ten Dijke1,2 *
1 Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC, Leiden,
The Netherlands
2 Uppsala University and Ludwig Institute for Cancer Research, Box 595, 75124, Uppsala, Sweden

*Correspondence to: Peter ten Dijke, Department of Molecular Cell Biology and Centre for Biomedical Genetics, Leiden University Medical Center,
Building 2, Room R-02-022, Postzone S-1-P, Postbus 9600, 2300 RC, Leiden, The Netherlands e-mail: p.ten_dijke@lumc.nl

Abstract
The transforming growth factor-β (TGF-β) signalling pathway plays a critical and dual role in the progression of
human cancer. During the early phase of tumour progression, TGF-β acts as a tumour suppressor, exemplified
by deletions or mutations in the core components of the TGF-β signalling pathway. On the contrary, TGF-β also
promotes processes that support tumour progression such as tumour cell invasion, dissemination, and immune
evasion. Consequently, the functional outcome of the TGF-β response is strongly context-dependent including
cell, tissue, and cancer type. In this review, we describe the molecular signalling pathways employed by TGF-β
in cancer and how these, when perturbed, may lead to the development of cancer. Concomitantly with our
increased appreciation of the molecular mechanisms that govern TGF-β signalling, the potential to therapeutically
target specific oncogenic sub-arms of the TGF-β pathway increases. Indeed, clinical trials with systemic TGF-β
signalling inhibitors for treatment of cancer patients have been initiated. However, considering the important
role of TGF-β in cardiovascular and many other tissues, careful screening of patients is warranted to minimize
unwanted on-target side effects.
Copyright  2010 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Keywords: BMP; cancer; epithelial to mesenchymal transition; EMT; metastasis; signal transduction; Smad; TGF-β; ubiquitin

Received 3 August 2010; Revised 18 August 2010; Accepted 1 September 2010

No conflicts of interest were declared.

Introduction pre-clinical models [6,7]. Nonetheless, issues with dos-


ing and timing of TGF-β treatment, among others,
Transforming growth factor-β (TGF-β) emerged with halted the translation of these findings towards clinical
the evolution of multi-cellular organisms, where it application.
plays an essential role in the development of the Importantly, analysis of human tumour samples
body plan during embryogenesis and is crucial for tis- has also suggested an active role for the TGF-β
sue homeostasis. The TGF-β pathway mediates such pathway in tumour progression. Immuno-staining for
regulation through control of proliferation, differentia- TGF-β correlated with metastasis in breast, colon,
tion, apoptosis, adhesion, invasion, and cellular micro- and prostate cancer [8–10]. In addition, the intensity
environment [1–4]. Consequently, malfunctioning of of TGF-β staining in invading lymph node metas-
this pathway has adverse effects and inactivation of tases was higher in breast and colon cancers than
components in this signalling pathway is central to in the primary tumour [11,12]. Moreover, TGF-β
many diseases including the development of tumouri- increases the motility and invasion of certain can-
genesis and tumour progression. In several types of cer cells, demonstrating that these cells, while hav-
human carcinomas, mutations or loss of heterozy-
ing lost sensitivity for TGF-β-induced growth arrest,
gosity (LOH) in central components of the TGF-β
remained TGF-β-responsive [13]. Furthermore, TGF-
pathway has been observed [5]. These observations
β secreted by tumour cells stimulates stroma forma-
support the idea that the TGF-β pathway has a tumour-
suppressive role that needs circumvention to allow tion and immune evasion of tumour cells [14,15].
tumourigenesis. Also, due to its growth-suppressive Thus, TGF-β has both tumour-suppressive and tumour-
effects, in the past TGF-β itself has been regarded promoting functions [16]. In this review, we will
as an attractive cytokine for the treatment of can- elaborate on the dynamic role that TGF-β plays in
cer. Therefore, studies were initiated in which TGF-β cancer biology; how perturbation of the TGF-β sig-
was explored as an adjuvant for chemotherapy. Indeed, nalling pathways contributes to cancer; and how new
TGF-β was able to protect normal cells and sensitize insights provide opportunities for targeted therapy of
tumour cells towards standard chemotherapy in some cancer patients.
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
206 E Meulmeester and P ten Dijke

Molecular mechanism of TGF-β signalling intense regulation [33]. Presentation and sequestration
of R-Smads to TGF-β receptor is controlled by SARA
and TMEPAI, respectively [34,35]. Phosphorylation
Canonical signalling via Smad proteins
of Smad proteins is reversed by phosphatases (eg
TGF-β is the founding member of a larger family PPM1A, PDP, and SCP1, 2 and 3), thereby creating
of secreted dimeric cytokines that comprise TGF- a rapid activation–deactivation cycle [36–38]. Fur-
βs, activins, bone morphogenetic proteins (BMPs), thermore, activated Smad2/3 proteins are counteracted
and growth and differentiation factors (GDFs) [4]. In by ubiquitin E3 ligases that target them for proteaso-
mammals, three alternative TGF-β isoforms exist, ie mal degradation [39,40]. In addition, the interaction
TGF-β1, TGF-β2, and TGF-β3. The bioactive ligands between phosphorylated Smad2 and Smad4 is impeded
are composed of homo- or hetero-dimers of polypep- by mono-ubiquitination of Smad4. Ectodermin is a
tides that are synthesized as precursor molecules and ubiquitin E3 ligase that targets Smad4 for mono-
matured by proteolytic cleavage by endoproteases such ubiquitination, which is counteracted by the deubiqui-
as Furin [17–19]. The cleaved N-terminal peptide, also tinating enzyme USP9x [41]. Moreover, the inhibitory
known as latency-associated peptide (LAP), forms a Smads (I-Smads), Smad6 and Smad7, are transcription-
non-covalent interaction with the C-terminal peptide ally induced upon BMP and TGF-β signalling [42–45].
dimer (TGF-β). Subsequent binding to the latent TGF- While Smad6 mainly inhibits BMP signalling, Smad7
β-binding protein (LTBP) allows the formation of the has the capability to prevent both BMP and TGF-β
large latency complex (LLC) that facilitates secretion signalling. Mechanistically, it has been proposed that
into the extracellular matrix [20]. By release from this I-Smads repress signalling by competing with R-Smads
complex, TGF-β is activated. for receptor binding [43,44,46]. Secondly, it was shown
Active TGF-β dimers mediate signalling through the that overexpression of Smad6 prevents the formation of
TGF-β type I and type II receptors (TGF-βRI and Smad1–Smad4 complexes, by binding to Smad1 [47].
TGF-βRII, respectively) that are endowed with ser- Smad7 has been implicated in the recruitment of the
ine/threonine kinase activity [21–23]. The membrane- E3 ubiquitin ligases Smurf1 and Smurf2 to activated
anchored proteoglycan betaglycan (TGF-βRIII) assists TGF-β receptors, which in turn leads to their degra-
TGF-β binding to TGF-βRII [24]. High affinity bind- dation via the ubiquitin proteasome system [48–50].
ing of TGF-β to TGF-βRII leads to heterotetrameric The importance of Smad7-mediated inhibition of TGF-
complex formation with TGF-βRI, which results in the β signalling is underscored by the observation that
phosphorylation of TGF-βRI by TGF-βRII (Figure 1). Smad7 is overexpressed in endometrial carcinomas and
In most cell types, TGF-βRI (also known as activin thyroid follicular cell lines [51,52]. Another point of
receptor-like kinase 5; ALK5) transduces signalling, regulation of TGF-β signalling is by the transcriptional
while in certain cell types ALK1 or other type I recep- repressors Ski, SnoN, and members of the HDAC fam-
tors can mediate signalling responses [25,26]. The type ily that interact with Smad2/3 and Smad4 [53–56].
I receptor propagates signalling by recruitment and Even though Smad signalling is required for the
phosphorylation of receptor-regulated Smad (R-Smad) majority of TGF-β-mediated signalling, not all re-
proteins. Whereas ALK5 signalling is mediated by sponses to TGF-β are solely dependent on Smad4
phosphorylation of Smad2 and Smad3 proteins, ALK1 [57,58]. For example, transcriptional intermediary fac-
signalling is mediated by Smad1, Smad5, and Smad8. tor 1γ (TIF1γ) selectively binds receptor-phosphory-
Activated Smads form a complex with the common lated Smad2/3 in competition with Smad4, in the
Smad (co-Smad; Smad4 in mammals) and shuttle into control of haematopoietic cell fate by TGF-β [59]. In
the nucleus [21,27]. Since activated Smad complexes addition, IKKα was identified as a critical co-regulator
have a weak binding affinity for DNA, additional of Smad2/3 in a Smad4-independent manner, which
DNA-binding transcription factors are required to reg- controls keratinocyte differentiation [60].
ulate high affinity interaction and specificity [28,29].
A variety of transcription factor families have been Non-canonical signalling
described to act in concert with Smad proteins such
as p300/CBP, Forkhead, homeobox, zinc-finger, AP1, While canonical signalling directly regulates the tran-
Ets, and basic helix-loop-helix families [30,31]. The scription of Smad-dependent target genes, Smad
diversity of R-Smad/co-Smad/co-factor combinations proteins have also been shown to participate in seques-
regulates the transcription of a vast amount of target tration, recruitment, and enzyme activation [61–63].
genes. The cell type-specific responses observed upon R-Smads (independent of Smad4) are also involved in
TGF-β stimulation can at least in part be explained by the regulation of miRNA maturation in the nucleus
the differential expression of these regulators in such [64]. Moreover, alternative signalling modules are
cell types [32]. present in parallel with Smad signalling that are
also responsive to TGF-β. The existence of Smad-
independent signalling is supported by the identifi-
Regulation of TGF-β Smad signalling cation of the TRAF6–TAK1–p38/JNK pathway as
To control the intensity and duration of TGF-β sig- a TGF-β signalling module downstream of TGF-β
nalling, each step in this signalling pathway is under receptors [65–67]. Furthermore, TGF-β signalling is
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 207

Figure 1. The TGF-β signalling pathway. Transforming growth factor-β (TGF-β) present in the extracellular matrix resides in the large
latent complex (LLC). Upon release, active TGF-β weakly interacts with the large betaglycan membrane protein that is present in vast
excess compared with TGF-βRII. Subsequently, TGF-β is presented to TGF-βRII, which leads to the formation of a heterotetrameric
complex between the serine/threonine kinases TGF-βRI and TGF-βRII. The constitutive active TGF-βRII phosphorylates (P) TGF-βRI, which
in turn recruits, phosphorylates, and activates Smad transcription factors (Smad2/3). Phosphorylated Smad2/3 form a complex with the
co-Smad (Smad4), translocate into the nucleus, and further build transcription complexes with additional co-repressors or co-activators to
regulate the expression of a wide variety of genes. The inhibitory Smad (Smad7) reduces further signalling by preventing phosphorylation
of Smad2/3. Besides Smad-mediated signalling, TGF-β also activates several other signalling cascades such as TRAF6–TAK1–p38/JNK,
RhoA–Rhock1, and Par6.

engaged in RhoA–Rock1 signalling that is required Mechanisms of TGF-β signalling in tumour


for the epithelial-to-mesenchymal transition (EMT) suppression
[68,69]. The TGF-β receptor also activates Erk-MAP
kinase signalling through direct phosphorylation of Shc In several cancers, deletion or mutations of TGF-β sig-
on tyrosine and serine residues by TGF-βRI [70]. TGF- nalling components alleviate the tumour-suppressive
βRII can also signal independently of TGF-βRI by effects of this pathway. In colon cancer with microsatel-
direct phosphorylation of, for example, Par6 that con- lite instability, TGF-βRII is subjected to accumulation
tributes to EMT by a loss of tight junctions [71]. While of replication errors, leading to inactivation of the
an increasing number of proteins have been identified receptor and thus the TGF-β pathway [74]. Similarly,
to interact with the TGF-β receptors [72,73], novel mutations in TGF-βRII have been described in gastric
mechanisms of Smad-independent signalling will most tumours, gliomas, and colorectal cancer [75,76]. While
likely be uncovered in the near future. mutations in TGF-βRI are less frequent, these have
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
208 E Meulmeester and P ten Dijke

been observed in pancreas, ovarian, and breast can- p15 (INK4B) and p21(WAF1), which are driven by
cer [77–79]. Downstream of the receptors, inactivating Smad3/Smad4 complexes in concert with FoxO and
mutations or deletions of SMAD4 are present in half Sp1 transcription factors [87–91]. p15 inhibits cell
of the pancreatic tumours and to a lesser extent in gas- cycle progression at the late G1 phase by interact-
trointestinal tumours [75,78]. However, tissue-specific ing with CDK4/6 and preventing their interaction with
inactivation of Smad4 or TGF-βRII alone in mouse cyclin D [92]. Consequently, the CDK inhibitor p27
models rarely leads to spontaneous tumour forma- is relocated from cyclin D–CDK4 complexes to inter-
tion, suggesting that the outcome of interference with act with and inhibit cyclin E–CDK2 complexes. p21
TGF-β signalling is strongly dependent on the context also inhibits the activity of cyclin E–CDK2 complexes
of individual tumours [80–85]. In fact, the tumour- [4,92]. The inactivity of these CDK complexes pre-
suppressive role of TGF-β is most apparent under vents phosphorylation of pRb, a major switch in cell
conditions of oncogenic stress. For example, inactiva- cycle progression, and thus progression through G1
tion of Smad4 or TGF-βRII in adenomatosis polyposis into S phase. While the induction of p21 and p15
coli (APC)-deficient mice potentiates the progression is important for the cytostatic effect in neuronal and
of intestinal polyps to carcinoma [83,86]. Similarly, epithelial cells, in haematopoietic cells TGF-β medi-
mammary tumours initiated by polyoma virus middle- ates its growth inhibition through the CDK inhibitor
T oncogene and premalignant lesions initiated by the p57 [93].
KRAS oncoprotein in skin, pancreas, and oral and Simultaneously with the activation of cell cycle pro-
oesophagus epithelium progress faster when TGF-β gression inhibitors, TGF-β represses the c-Myc onco-
signalling is crippled [80–82,84]. gene that promotes cell proliferation. c-Myc is a tran-
Mechanistically, TGF-β has several operating arms scription factor that activates or represses transcription
to achieve its tumour-suppressive effect which include depending on its target genes. Since c-Myc inhibits
regulation of cell proliferation, apoptosis, and indi- transcriptional activation of p15 and p21, through Miz-
rectly through the tumour stroma. 1, TGF-β has a firm grip on the regulation of these
target genes [94,95]. Furthermore, TGF-β represses the
Regulation of cell proliferation and apoptosis expression of Id1, Id2, and Id3, which are nuclear
In epithelial, neuronal, and haematopoietic cells, TGF- factors implicated in differentiation and progression
β limits cell proliferation through a coordinated pro- through the G1–S cell cycle transition [96]. More
gramme of cytostatic gene responses (Figure 2A). recently, TGF-β’s anti-proliferative effect was shown
At the core of these events is the induction of to be dependent on the eukaryotic translation initiation
cyclin-dependent kinase (CDK) inhibitors such as factor-4F (eIF4F) [97]. Thus, TGF-β orchestrates its

Figure 2. TGF-β as a tumour suppressor. (A) TGF-β controls cell cycle progression by repressing the oncogenic Myc transcription factor,
which in turn prevents transcriptional activation of the CDK inhibitors p21 and p15 via interaction with MIZ1. TGF-β also directly
transcriptionally activates p21 and p15. p15 abolishes the interaction of cyclin D (Cyc D) with the CDK4/6 complex, thereby inactivating
the CDK. p21 (and also p27) inactivates the cyclin E (Cyc E) CDK2 complex, which leads to stalling of the cell cycle at the G1–S boundary.
(B) TGF-β inhibits apoptosis in a variety of cell types; however, the exact molecular mechanisms remain to be determined. (C) Through
non-cell autonomous signalling, TGF-β controls tumour progression via inhibition of paracrine growth factors in the tumour stroma during
the early stages of tumour development. The absence of TGF-β signalling in tumour stroma cells relieves the growth inhibitory effect on
epithelial cells, which consequently obtain increased growth, migratory, and invasive properties.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 209

growth-suppressive effects via functionally redundant subsets of colon, pancreatic, and gastric cancers, muta-
mechanisms, ie by slowing down cell growth progres- tion or deletion of TGF-β receptors or Smads in
sion and simultaneously de-accelerating proliferative the TGF-β pathway leads to its inactivation or per-
actions. turbation of signalling responses [74–76,107–110].
In addition to the regulatory role of TGF-β in cell Tumours that acquire the ability to bypass the tumour-
cycle progression, tumour progression is also limited suppressive arms may exploit certain aspects of TGF-β
through control of the apoptotic response. TGF-β has signalling to actively promote tumour cell progression.
been reported to induce apoptosis in a variety of cell In fact, aggressive tumours resistant to the tumour-
types under physiological circumstances; however, the suppressive effects of TGF-β preserve the core compo-
molecular mechanisms remain ill defined (Figure 2B). nents of TGF-β signalling. Different types of tumours
Candidates that contribute to the apoptotic functions such as gliomas and breast and prostate cancer seem to
of TGF-β include the death receptor FAS, growth acquire mutations preferentially not in the core compo-
arrest and DNA damage inducible 45β (GADD45β), nents of TGF-β signalling [109,111–114]. Such human
BIM, and death-associated kinase (DAPK) [98–101]. tumours likely obtain resistance to TGF-β-mediated
Confirmation of the physiological relevance of these growth arrest and importantly retain the ability to
candidates awaits experimental proof using in vivo exploit TGF-β signalling to induce pathways that pro-
model systems. A further thorough understanding of mote EMT, tumour invasion, metastatic dissemination,
the mechanisms by which TGF-β exerts its cytostatic and evasion of the immune system. Therefore, tumours
and apoptotic responses is important to identify key with such a signature are highly aggressive.
points where tumours may hijack the TGF-β system to
favour tumour progression. Epithelial-to-mesenchymal transition (EMT)
and invasion
Indirect regulation of tumour suppression EMT is a key process during embryonic development
In addition to cell autonomous regulation of apoptosis that contributes to the formation of the body plan and
and growth arrest, TGF-β further restricts growth and allows differentiation of multiple tissues and organs.
tumour progression of epithelial cells through blockage Furthermore, it plays an important role during wound
of paracrine factor production in the tumour stroma healing, where keratinocytes recapitulate part of the
(Figure 2C). While the impact of the stromal fibrob- EMT process to acquire a more migratory character
last on tumour progression has been known since to seal the wounded area. Importantly, EMT also
early studies on breast cancer, the role of TGF-β in plays a pivotal role in pathological disorders such
this process emerged from mouse models in which as fibrosis and cancer progression. Even though the
TGF-β signalling was impaired in stromal fibroblasts EMT process is well accepted in a variety of cancer
[102,103]. Overexpression of a kinase inactive mutant cell models, the criticism on its relevance for human
of TGF-βRII in the mammary stromal cells results cancer progression has only recently dwindled, due
in epithelial hyperplasia and increased production of to convincing morphological evidence of EMT at
hepatocyte growth factor (HGF) [103]. These observa- the invasive fronts of human tumours [115–117]. To
tions were strengthened by a mouse model in which invade normal tissue and spread to distant organs,
TGF-βRII was conditionally inactivated in fibroblasts, carcinoma cells need to lose cell polarity, cell–cell
which results in the development of squamous cell car- contacts, and acquire fibroblastic-like properties. In
cinoma of the forestomach and prostatic intraepithelial this process of EMT, cells become highly motile
neoplasia [104]. The elevated expression of HGF in and invasive, which allows survival in an anchorage-
the TGF-βRII-deficient fibroblast led to the activation independent environment and provides them with stem
of its receptor (Met) in adjacent epithelial cells. Fur- cell-like properties (Figure 3). A molecular hallmark
thermore, the loss of TGF-βRII in fibroblasts results for cells undergoing EMT is decreased expression
in increased TGF-α and macrophage-stimulating pro- of epithelial cell–cell junction proteins, such as E-
tein (MSP), which all together contribute to augmented cadherin and zona occludens (ZO-1), while at the same
tumour growth, motility, and invasion [105]. Moreover, time acquiring the expression of mesenchymal markers,
in an Eµ-Myc transgenic mouse lymphoma model, such as vimentin, α-smooth muscle actin (α-SMA), and
TGF-β secreted from non-neoplastic macrophages fibronectin [1,118–121]. Furthermore, up-regulation
prevents tumour development by promoting cellular of matrix metalloproteases and N-cadherin led to
senescence [106]. the degradation of extracellular matrix proteins and
rendered tumour cells more migratory, respectively.
The identification of TGF-β as a major inducer of
EMT came from studies in cell culture. Treatment
Tumour-promoting roles of TGF-β signalling of normal mouse breast epithelial cells with TGF-β
changes the cuboidal shape to an elongated spindle,
During the early stages of tumour development, the accompanied by a decrease in epithelial markers and
TGF-β pathway operates as a tumour suppressor, increased expression of mesenchymal markers [122].
thereby preventing tumour growth. As seen in large Canonical TGF-β–Smad signalling plays a pivotal role
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
210 E Meulmeester and P ten Dijke

Figure 3. TGF-β as a tumour promoter. (A) TGF-β stimulates the progression of carcinoma in situ towards a more aggressive, motile, and
invasive carcinoma. The tumour cells (yellow) acquire the capacity to invade adjacent tissues and intravasate into the blood stream.
(B) TGF-β promotes metastatic dissemination of primary breast carcinoma towards bone by up-regulating PTHrP and IL11, which in turn
activate osteoblasts (green) to secrete RANKL. RANKL leads to the differentiation of precursor cells (blue) into osteoclasts (orange), which
absorb the bone mass and release stored TGF-β. As such, a feed-forward loop is created in which TGF-β promotes the growth of bone
metastasis. (C) By inducing ANGPTL4, TGF-β primes tumour cells for dissemination towards the lungs. ANGPTL4 enhances extravasation
by dissociating vascular endothelial cell–cell junctions. TGF-β, released by the tumour cells, allows evasion of the immune response by
inactivating CD4+ and CD8+ T-cells that normally inhibit the growth of tumour cells.

in this process, as depletion of Smad3/Smad4 or over- palatal shelves, a process that is dependent on EMT.
expression of Smad7 completely abolishes induction Consequently, knockout mice for TGF-β3 are con-
of EMT [123–125]. Additionally, increased expres- fronted with a cleft palate [128,129]. Furthermore,
sion of Smad3 and Smad4 in the presence of con- TGF-β2 knockouts suffer from a wide variety of devel-
stitutive active TGF-βRI enhances induction of EMT opmental defects that can at least in part be explained
[126]. Mechanistically, TGF-β orchestrates a diverse by a dysfunctional EMT process [130]. Additionally,
transcriptional network that involves transcriptional in a mouse model for skin carcinogenesis, increased
activation of Snail, ZEB, and the BHLH family of expression of TGF-β1 in keratinocytes enhances EMT
proteins [119]. On the other hand, TGF-β mediates and increases the rate and aggressiveness of these
down-regulation of the microRNA-200 family that is tumours, induced by chemical carcinogenesis [131]. In
required for epithelial cells to undergo EMT [127]. this model, keratinocyte-specific depletion of Smad2
The expression of these factors is regulated by both stimulates EMT by promoting Smad3/Smad4-mediated
Smad-dependent and Smad-independent mechanisms Snail transcription, indicating that Smad2 and Smad3
that lead to the repression of epithelial cell markers have differential functions [132]. Evidence for TGF-
and induction of mesenchymal genes. β as a regulator of EMT in human cancer has been
Shortly after the identification of TGF-β as a reg- suggested by Shipitsin et al, who describe an acti-
ulator of EMT in cell culture conditions, work from vated TGF-β pathway in isolated CD44-positive can-
mouse models confirmed TGF-β as a critical regulator cerous cells compared with normal tissue [133]. It
of EMT in vivo. During normal craniofacial develop- appears that during the TGF-β-induced EMT process,
ment, TGF-β3 is required for the fusion of the two epithelial cells also acquire stem cell-like properties
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 211

besides obtaining a mesenchymal phenotype [134]. hormone-related protein (PTHrP) [146,147]. The post-
Interestingly, Ikushima et al recently reported mech- translationally increased levels of PTHrP subsequently
anistic insights into how TGF-β maintains stemness in stimulate production of RANK ligand (RANKL) in
glioma initiating cells [135]. TGF-β directly regulates osteoblasts, which in turn promotes the differenti-
the expression of Sox4, which in turn mediates the ation of osteoclast precursors and bone resorption
induction of Sox2 expression [135]. (Figure 3B) [148,149]. Furthermore, it was recently
demonstrated that TGF-βRII also directly phosphory-
lates the PTH receptor and via this route could also
Metastatic dissemination contribute to bone resorption [72].
After invasion into adjacent tissue, the metastatic Transcriptome comparison, of adrenal with bone
process continues through intravasation, dissemination metastasis, originated from a breast cancer epithelial
to distal capillary beds, extravasation, and growth in a cell line inoculated into mice, led to the identification
distal organ [136,137]. Particular tumour types have a of CTGF and IL-11 within a larger gene signature as
tendency to metastasize to certain organs; for example, critical factors for bone metastasis [138]. The TGF-
breast tumours tend to metastasize towards the brain, β-mediated induction of IL-11 and CTGF, as well
lungs, bones, and liver. This distribution pattern seems as the acquired metastatic capacity, requires Smad4-
to be dependent on the expression of a specific set of dependent signalling [123,150]. Besides Smad4, Smad3
genes rather than vasculature, blood flow, and number is required for the induction of key target genes
of cells delivered to the receiving organ [138–140]. in metastasis [151]. Importantly, depletion of Smad2
The role of TGF-β in the metastatic process became enhanced the metastatic process of MDA-MB-231
evident with the observation that TGF-β immuno- cells, while Smad3 prevented the metastatic spread
staining was much stronger in metastasized breast [151].
tumours than in the primary tumour [11]. In addition, Besides stimulating metastatic colonization, TGF-β
expression of TGF-βRII has a negative correlation with also has the ability to prime tumour cells for metastatic
overall survival in oestrogen receptor (ER)-negative dissemination. By employing a cell culture-derived
breast cancer patients [141]. Importantly, chemother- TGF-β gene response signature, angiopoietin-like 4
apy or radiation treatment in the MMTV/PyVmT trans- (ANGPTL4) was identified as a key player to prime
genic model of breast metastasis led to increased TGF- breast cancer cells for metastasis towards the lungs
β1 levels as well as increased circulating tumour cells [152]. ANGPTL4 assists the tumour cells to disrupt
and lung metastasis [142]. Administration of neutraliz- the lung capillaries and thereby enables pulmonary
ing TGF-β antibodies prevents this increase in metasta- metastasis (Figure 3C). In a previous study ANGPLT4
sis, thus suggesting an important role for TGF-β in the has also been identified within a TGF-β-responsive
metastatic process [142]. However, the role of TGF-β gene signature that mediates metastasis towards the
in metastasis progression seems to be strongly context- lungs [139]. The presence of dominant negative TGF-
dependent. For example, expression of an activated βRI or the absence of Smad4 in ER-negative breast
TGF-βRI transgene increases metastasis to the lungs, cancer cells prevented their capacity to metastasize
while targeted deletion of TGF-βRII resulted in a sim- when implanted as mammary tumours.
These studies shed light on a role for TGF-β in
ilar observation [80,143,144]. The picture of TGF-β
the metastatic process, both in the early invasion and
as a promoter of metastasis is even further obscured
intravasation stage and during the later process involv-
by the observation that short-term stimulation with
ing extravasation and colony formation. However, most
TGF-β stimulates metastasis formation, while persis-
of these studies rely on either inoculation of cell
tent stimulation decreases the metastatic spread to the
lines or transplantation of tumours. For future work,
lungs [145]. From this study, it has been proposed that
it would be important to study these phenomena in a
TGF-β signalling initially needs to be high in order
more physiological setting employing mouse models in
to acquire invasive properties for dissemination, while
which metastases arise spontaneously from the primary
upon extravasation TGF-β signalling is low to allow
tumour. In such a setting, the role of TGF-β in the var-
proliferation at the secondary site.
ious metastatic processes for different types of cancer
Insights into the mechanism by which TGF-β stim-
could be investigated employing inducible knockout or
ulates metastatic dissemination came from studies on
knock-in models.
bone and lung metastasis. Primary breast and prostate
tumours have in common that they often metastasize
towards bone, a process in which TGF-β plays an Immune evasion
important role upon the arrival of tumour cells. The In addition to regulation of EMT, invasion, and
presence of metastatic tumour cells in the bone micro- metastatic dissemination, TGF-β also supports tumour
environment leads to activation of osteoclasts, which progression by evading the immune system [2]. The
degrade the bone matrix and consequently release first evidence for TGF-β in immune evasion came
stored TGF-β and other growth factors. TGF-β in from the observation that TGF-β potently inhibits
turn stimulates the metastatic cells to release oste- tumour-induced CD8+ cytolytic T-lymphocyte (CTL)-
olytic cytokines such as, amongst others, parathyroid mediated rejection of a murine tumour [153]. Further
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
212 E Meulmeester and P ten Dijke

support for TGF-β as an immuno-suppressant came the production of the ectopic TGF-β is by using anti-
from a transgenic mouse model expressing a dom- sense molecules, which block TGF-β-mediated gene
inant negative form of TGF-βRII (dn-TGF-βRII) in expression. Antisense molecules for TGF-β1 demon-
all T-cells [154]. The absence of TGF-β signalling in strate promising results in pre-clinical experiments
T-lymphocytes suppressed growth and metastasis in by decreasing tumourigenicity [163,164]. A promising
this mouse model when challenged with melanoma candidate in further development is AP 12 009 (Anti-
or lymphoma cell lines. By employing differential sense Pharma), an antisense molecule directed against
immuno-depletion in these mice, it was determined that TGF-β2, which reduces TGF-β2-mediated migration
both T-helper (CD4+ ) and cytolytic T-cells (CD8+ ) and proliferation, as well as alleviating its immuno-
were responsible for tumour resistance. In addition, suppressive effects [165,166]. The efficacy in phase
inactivation of TGF-β signalling in CTLs (overexpres- I/II clinical trials to treat refractory high-grade glioma
sion of dn-TGF-βRII) in a mouse model for sponta- patients exceeded the expected median survival com-
neous prostate cancer delays tumour progression [155]. pared with chemotherapy [166]. These results suggest
Mechanistically, it was demonstrated that TGF-β tran- AP 12 009 as a promising therapeutic target to inhibit
scriptionally represses the production of several pro- TGF-β signalling for malignant tumour therapy. Cur-
apoptotic factors in CTLs such as perforin, granzyme rently, AP 12 009 is in further clinical studies to test
A, granzyme B, FAS ligand, and interferon-γ [156]. its efficacy for metastatic melanoma, pancreatic carci-
In addition to its immuno-suppressive effect on CTLs, noma, and metastatic colorectal carcinoma.
TGF-β also represses the activity of natural killer (NK)
cells. Inhibition of TGF-β using neutralizing TGF-β Ligand traps that sequester TGF-β
antibodies increased NK cell activity and consequently Another approach to prevent TGF-β signalling is
resulted in the suppression of metastasis formation trapping the TGF-β ligand employing soluble TGF-
of an inoculated breast carcinoma cell line [157]. β receptors or neutralizing antibodies. Soluble TGF-
CD4+ CD25+ regulatory T-cells that produce high βRII and TGF-βRIII have proven their value in
amounts of TGF-β take part in the inhibition of NK cell pre-clinical model systems. For example, expression
activity [158]. TGF-β inhibits NK-mediated cytotoxic- of soluble TGF-βRII prevents pulmonary, pancreatic
ity through transcriptional repression of the activating or liver metastasis in mouse models [167–169], while
receptor NKG2D and the type I transmembrane pro- intraperitoneal delivery of the soluble ectodomain of
tein NKp30 [159,160]. Neutralizing antibodies towards TGF-βRIII into athymic nude mice inhibited pul-
TGF-β could restore the NKG2D expression, as well monary metastasis [170]. Alternatively, TGF-β neutral-
as the anti-tumour reactivity [161]. In glioblastoma izing antibodies such as 2G7 and 1D11 prevent the
patients, TGF-β decreases the expression of NKG2D interaction of TGF-β with its receptors and thereby
in CTLs and NK cells, and represses the expression of restrain its biological activity [157,171]. Administra-
MICA (the ligand of NKG2D) in glioma cells [162]. tion of 1D11 following inoculation of 4T1 cells sup-
pressed pulmonary metastasis, while 2G7 transiently
reduced the growth rate of intra-abdominal transplanted
Therapeutic exploitment of the TGF-β pathway MDA-MB-231 cells and lung metastasis [157,172].
Based on these results, Genzyme developed a mon-
oclonal neutralizing TGF-β antibody (GC1008) for
A rationale for designing therapeutic inhibitors that which recently a clinical phase I/II trial was com-
block TGF-β signalling in human cancer was initiated pleted to assess the safety and efficacy in patients with
by the observation that excess TGF-β promotes tumour advanced metastatic melanoma or renal cell carcinoma.
progression. The current strategies to target TGF-β
mainly focus on general inhibition of TGF-β signalling,
Small molecule inhibitors of TGF-β signalling
which can be subdivided into three major approaches.
Firstly, the synthesis of TGF-β can be prevented by Additionally, small molecule inhibitors that restrain the
administration of antisense molecules. Secondly, ligand kinase activity of TGF-β receptors can prevent TGF-β
traps (including monoclonal TGF-β neutralizing anti- signalling. The compound SB-431 542 prevents TGF-
bodies and soluble TGF-βRII or TGF-βRIII) prevent β-mediated Smad2/3 phosphorylation by restraining the
soluble TGF-β from activating its signalling cascade. kinase activity of TGF-βRI [173]. Treatment of human
Thirdly, small molecule inhibitors hinder the kinase osteosarcoma cells with SB-431 542 inhibits TGF-β-
activity of TGF-β receptors. induced proliferation, while in malignant glioma cells
angiogenesis, proliferation, and motility are inhibited
[174,175]. Another inhibitor of TGF-βRI is SD-208,
Targeting TGF-β signalling using antisense which upon systemic administration increases median
molecules survival upon implantation of malignant glioma cells
During tumour progression, the production of TGF-β into the mouse brain [176]. In another study, SD-208
is often enhanced either by fibroblasts in the stroma or was reported to inhibit TGF-β-induced EMT, migra-
by the tumour itself, which correlates with increased tion, and invasion of two murine breast carcinoma cell
aggressiveness of the tumour. One strategy to diminish lines, while also inhibiting the metastasis formation of
Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 213

these cell lines when inoculated into the mammary fat Teaching materials
pads of mice [177]. The compound Ki26894, also a
TGF-βRI kinase inhibitor, prevents the motility and
PowerPoint slides of the figures from this review
invasion of breast and gastric cancer cells [178,179].
are supplied as supporting information in the online
Furthermore, LY2 109 761 is a small molecule that
version of this article.
inhibits the kinase activity of both TGF-βRI and TGF-
βRII, and has been shown to inhibit the metastatic
spread to a variety of organs [180,181]. The small References
molecule compound LY2 157 299 (Eli Lilly & Co) is
currently in a clinical phase I trial to address its safety 1. Heldin CH, Landstrom M, Moustakas A. Mechanism of TGF-β
and pharmacokinetics. LY2 157 299 has been reported signaling to growth arrest, apoptosis, and epithelial–mesenchymal
in mouse studies to inhibit the primary tumour growth transition. Curr Opin Cell Biol 2009; 21: 166–176.
of a breast and lung cancer cell line [182]. 2. Yang L, Pang Y, Moses HL. TGF-β and immune cells: an impor-
tant regulatory axis in the tumor microenvironment and progres-
sion. Trends Immunol 2010; 31: 220–227.
Conclusion and future perspectives 3. Ikushima H, Miyazono K. TGFβ signalling: a complex web in
cancer progression. Nature Rev Cancer 2010; 10: 415–424.
4. Massague J. TGFβ in Cancer. Cell 2008; 134: 215–230.
With the continuously increasing appreciation of the
5. Levy L, Hill CS. Alterations in components of the TGF-β super-
clinical importance of TGF-β as a tumour promoter,
family signaling pathways in human cancer. Cytokine Growth
interest in targeting the TGF-β pathway for therapeutic Factor Rev 2006; 17: 41–58.
intervention is rising. The use of genetic screens that 6. Sonis ST, Van Vugt AG, Brien JP, et al . Transforming growth
delineate the tumour-suppressive versus the tumour- factor-β3 mediated modulation of cell cycling and attenuation
promoting roles of TGF-β signalling will provide a of 5-fluorouracil induced oral mucositis. Oral Oncol 1997; 33:
basis for new research that will enable the target- 47–54.
ing of its specific oncogenic sub-arms. Several pre- 7. Goey H, Keller JR, Back T, et al . Inhibition of early murine
clinical studies led to clinical trials that underline the hemopoietic progenitor cell proliferation after in vivo locoregional
potential to restrain TGF-β signalling [183,184]. Fur- administration of transforming growth factor-β1. J Immunol 1989;
thermore, treatment using soluble TGF-βRII prevents 143: 877–880.
the metastatic spread in a mouse model, while severe 8. Wikstrom P, Stattin P, Franck-Lissbrant I, et al . Transforming
pathological disorders observed as in TGF-β knockout growth factor β1 is associated with angiogenesis, metastasis,
and poor clinical outcome in prostate cancer. Prostate 1998; 37:
mice remain absent [168]. However, some concerns
19–29.
regarding the general systemic inhibition of TGF-β
9. Walker RA, Dearing SJ. Transforming growth factor β1 in ductal
remain, due to its tumour-suppressive role [74–79]. carcinoma in situ and invasive carcinomas of the breast. Eur
Furthermore, TGF-β also has a profound role in the car- J Cancer 1992; 28: 641–644.
diovascular system, such that general TGF-β inhibitors 10. Friedman E, Gold LI, Klimstra D, et al . High levels of trans-
will most likely have adverse on-target side effects. forming growth factor β1 correlate with disease progression in
Therefore, to determine the treatment regimen, it is of human colon cancer. Cancer Epidemiol Biomarkers Prev 1995;
critical importance to define and select those patients 4: 549–554.
who are confronted with tumours in which TGF-β’s 11. Dalal BI, Keown PA, Greenberg AH. Immunocytochemical local-
tumour-promoting role prevails. Although individual- ization of secreted transforming growth factor-β1 to the advancing
based medicine still has a long way to go, the current edges of primary tumors and to lymph node metastases of human
state-of-the-art techniques such as microarray, pro- mammary carcinoma. Am J Pathol 1993; 143: 381–389.
tein microarray or super-SILAC pave the way for a 12. Picon A, Gold LI, Wang J, et al . A subset of metastatic human
colon cancers expresses elevated levels of transforming growth
detailed view of key corrupted pathways in individuals
factor β1. Cancer Epidemiol Biomarkers Prev 1998; 7: 497–504.
[185–187].
13. ten Dijke P, Goumans MJ, Itoh F, et al . Regulation of cell
proliferation by Smad proteins. J Cell Physiol 2002; 191: 1–16.
Acknowledgment 14. Ikushima H, Miyazono K. Cellular context-dependent ‘colors’ of
We are grateful to all laboratory members for stimulat- transforming growth factor-β signaling. Cancer Sci 2010; 101:
ing discussions. We apologize to those whose work 306–312.
we could not cite due to limited space constraints. 15. Flavell RA, Sanjabi S, Wrzesinski SH, et al . The polarization of
immune cells in the tumour environment by TGFβ. Nature Rev
Our work is supported by grants from the LUMC
Immunol 2010; 10: 554–567.
(‘vrije beleidsruimte’), the Dutch Cancer Society, the
16. Roberts AB, Wakefield LM. The two faces of transforming
Netherlands Organization for Health and Development, growth factor β in carcinogenesis. Proc Natl Acad Sci U S A 2003;
the Swedish Cancer Foundation, and the Centre for 100: 8621–8623.
Biomedical Genetics. 17. Dubois CM, Laprise MH, Blanchette F, et al . Processing of
transforming growth factor β1 precursor by human furin conver-
tase. J Biol Chem 1995; 270: 10618–10624.
Author contribution statement 18. Cheifetz S, Weatherbee JA, Tsang ML, et al . The transforming
growth factor-β system, a complex pattern of cross-reactive
EM wrote the manuscript under the supervision of PtD. ligands and receptors. Cell 1987; 48: 409–415.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
214 E Meulmeester and P ten Dijke

19. Ling N, Ying SY, Ueno N, et al . Pituitary FSH is released by 42. Afrakhte M, Moren A, Jossan S, et al . Induction of inhibitory
a heterodimer of the β-subunits from the two forms of inhibin. Smad6 and Smad7 mRNA by TGF-β family members. Biochem
Nature 1986; 321: 779–782. Biophys Res Commun 1998; 249: 505–511.
20. Janssens K, ten Dijke P, Janssens S, et al . Transforming growth 43. Nakao A, Afrakhte M, Moren A, et al . Identification of Smad7,
factor-β1 to the bone. Endocr Rev 2005; 26: 743–774. a TGFβ-inducible antagonist of TGF-β signalling. Nature 1997;
21. Shi Y, Massagué J. Mechanisms of TGF-β signaling from cell 389: 631–635.
membrane to the nucleus. Cell 2003; 113: 685–700. 44. Imamura T, Takase M, Nishihara A, et al . Smad6 inhibits sig-
22. Heldin CH, Miyazono K, ten Dijke P. TGF-β signalling from cell nalling by the TGF-β superfamily. Nature 1997; 389:
membrane to nucleus through SMAD proteins. Nature 1997; 390: 622–626.
465–471. 45. Takase M, Imamura T, Sampath TK, et al . Induction of Smad6
23. Feng XH, Derynck R. Specificity and versatility in TGF-β signal- mRNA by bone morphogenetic proteins. Biochem Biophys Res
ing through Smads. Annu Rev Cell Dev Biol 2005; 21: 659–693. Commun 1998; 244: 26–29.
24. Lopez-Casillas F, Wrana JL, Massague J. Betaglycan pres- 46. Hayashi H, Abdollah S, Qiu Y, et al . The MAD-related protein
ents ligand to the TGFβ signaling receptor. Cell 1993; 73: Smad7 associates with the TGFβ receptor and functions as an
1435–1444. antagonist of TGFβ signaling. Cell 1997; 89: 1165–1173.
25. Goumans MJ, Valdimarsdottir G, Itoh S, et al . Activin receptor- 47. Hata A, Lagna G, Massague J, et al . Smad6 inhibits BMP/Smad1
like kinase (ALK)1 is an antagonistic mediator of lateral signaling by specifically competing with the Smad4 tumor sup-
TGFβ/ALK5 signaling. Mol Cell 2003; 12: 817–828. pressor. Genes Dev 1998; 12: 186–197.
26. Daly AC, Randall RA, Hill CS. Transforming growth factor 48. Kavsak P, Rasmussen RK, Causing CG, et al . Smad7 binds to
β-induced Smad1/5 phosphorylation in epithelial cells is medi- Smurf2 to form an E3 ubiquitin ligase that targets the TGF β
ated by novel receptor complexes and is essential for anchorage- receptor for degradation. Mol Cell 2000; 6: 1365–1375.
independent growth. Mol Cell Biol 2008; 28: 6889–6902. 49. Ebisawa T, Fukuchi M, Murakami G, et al . Smurf1 interacts with
27. Hill CS. Nucleocytoplasmic shuttling of Smad proteins. Cell Res transforming growth factor-β type I receptor through Smad7
2009; 19: 36–46. and induces receptor degradation. J Biol Chem 2001; 276:
28. Shi Y, Wang YF, Jayaraman L, et al . Crystal structure of a Smad 12477–12480.
MH1 domain bound to DNA: insights on DNA binding in TGF-β 50. Itoh S, Landström M, Hermansson A, et al . Transforming growth
signaling. Cell 1998; 94: 585–594. factor β1 induces nuclear export of inhibitory Smad7. J Biol Chem
29. Derynck R, Zhang Y, Feng XH. Smads: transcriptional activators 1998; 273: 29195–29201.
of TGF-β responses. Cell 1998; 95: 737–740. 51. Dowdy SC, Mariani A, Reinholz MM, et al . Overexpression of
30. Koinuma D, Tsutsumi S, Kamimura N, et al . Chromatin im- the TGF-β antagonist Smad7 in endometrial cancer. Gynecol
munoprecipitation on microarray analysis of Smad2/3 binding Oncol 2005; 96: 368–373.
sites reveals roles of ETS1 and TFAP2A in transforming growth 52. Cerutti JM, Ebina KN, Matsuo SE, et al . Expression of Smad4
factor β signaling. Mol Cell Biol 2009; 29: 172–186. and Smad7 in human thyroid follicular carcinoma cell lines.
31. Koinuma D, Tsutsumi S, Kamimura N, et al . Promoter-wide J Endocrinol Invest 2003; 26: 516–521.
analysis of Smad4 binding sites in human epithelial cells. Cancer 53. Stroschein SL, Wang W, Zhou S, et al . Negative feedback regu-
Sci 2009; 100: 2133–2142. lation of TGF-β signaling by the SnoN oncoprotein. Science 1999;
32. Itoh S, Itoh F, Goumans MJ, et al . Signaling of transforming 286: 771–774.
growth factor-β family members through Smad proteins. Eur 54. Luo K, Stroschein SL, Wang W, et al . The Ski oncoprotein
J Biochem 2000; 267: 6954–6967. interacts with the Smad proteins to repress TGFβ signaling. Genes
33. Itoh S, ten Dijke P. Negative regulation of TGF-β receptor/Smad Dev 1999; 13: 2196–2206.
signal transduction. Curr Opin Cell Biol 2007; 19: 176–184. 55. Nomura T, Khan MM, Kaul SC, et al . Ski is a component
34. Tsukazaki T, Chiang TA, Davison AF, et al . SARA, a FYVE of the histone deacetylase complex required for transcriptional
domain protein that recruits Smad2 to the TGFβ receptor. Cell repression by Mad and thyroid hormone receptor. Genes Dev
1998; 95: 779–791. 1999; 13: 412–423.
35. Watanabe Y, Itoh S, Goto T, et al . TMEPAI, a transmembrane 56. Akiyoshi S, Inoue H, Hanai J, et al . c-Ski acts as a transcrip-
TGF-β-inducible protein, sequesters Smad proteins from active tional co-repressor in transforming growth factor-β signaling
participation in TGF-β signaling. Mol Cell 2010; 37: 123–134. through interaction with smads. J Biol Chem 1999; 274:
36. Lin X, Duan X, Liang YY, et al . PPM1A functions as a Smad 35269–35277.
phosphatase to terminate TGFβ signaling. Cell 2006; 125: 57. Subramanian G, Schwarz RE, Higgins L, et al . Targeting endoge-
915–928. nous transforming growth factor β receptor signaling in SMAD4-
37. Sapkota G, Knockaert M, Alarcon C, et al . Dephosphorylation of deficient human pancreatic carcinoma cells inhibits their invasive
the linker regions of Smad1 and Smad2/3 by small C-terminal phenotype1. Cancer Res 2004; 64: 5200–5211.
domain phosphatases has distinct outcomes for bone morpho- 58. Wisotzkey RG, Mehra A, Sutherland DJ, et al . Medea is a
genetic protein and transforming growth factor-β pathways. J Biol Drosophila Smad4 homolog that is differentially required to
Chem 2006; 281: 40412–40419. potentiate DPP responses. Development 1998; 125: 1433–1445.
38. Chen HB, Shen J, Ip YT, et al . Identification of phosphatases for 59. He W, Dorn DC, Erdjument-Bromage H, et al . Hematopoiesis
Smad in the BMP/DPP pathway. Genes Dev 2006; 20: 648–653. controlled by distinct TIF1γ and Smad4 branches of the TGFβ
39. Lo RS, Massagué J. Ubiquitin-dependent degradation of TGF-β- pathway. Cell 2006; 125: 929–941.
activated smad2. Nature Cell Biol 1999; 1: 472–478. 60. Descargues P, Sil AK, Sano Y, et al . IKKα is a critical coregu-
40. Gao S, Alarcon C, Sapkota G, et al . Ubiquitin ligase Nedd4L lator of a Smad4-independent TGFβ-Smad2/3 signaling pathway
targets activated Smad2/3 to limit TGF-β signaling. Mol Cell that controls keratinocyte differentiation. Proc Natl Acad Sci U S
2009; 36: 457–468. A 2008; 105: 2487–2492.
41. Dupont S, Mamidi A, Cordenonsi M, et al . FAM/USP9x, a deu- 61. Bonni S, Wang HR, Causing CG, et al . TGF-β induces assembly
biquitinating enzyme essential for TGFβ signaling, controls of a Smad2–Smurf2 ubiquitin ligase complex that targets SnoN
Smad4 monoubiquitination. Cell 2009; 136: 123–135. for degradation. Nature Cell Biol 2001; 3: 587–595.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 215

62. Conery AR, Cao Y, Thompson EA, et al . Akt interacts directly 82. Ijichi H, Chytil A, Gorska AE, et al . Aggressive pancreatic
with Smad3 to regulate the sensitivity to TGF-β induced apopto- ductal adenocarcinoma in mice caused by pancreas-specific
sis. Nature Cell Biol 2004; 6: 366–372. blockade of transforming growth factor-β signaling in coop-
63. Zhang L, Duan CJ, Binkley C, et al . A transforming growth eration with active Kras expression. Genes Dev 2006; 20:
factor β-induced Smad3/Smad4 complex directly activates protein 3147–3160.
kinase A. Mol Cell Biol 2004; 24: 2169–2180. 83. Munoz NM, Upton M, Rojas A, et al . Transforming growth fac-
64. Hata A, Davis BN. Control of microRNA biogenesis by TGFβ tor β receptor type II inactivation induces the malignant transfor-
signaling pathway—a novel role of Smads in the nucleus. mation of intestinal neoplasms initiated by Apc mutation. Cancer
Cytokine Growth Factor Rev 2009; 20: 517–521. Res 2006; 66: 9837–9844.
65. Ohkawara B, Shirakabe K, Hyodo-Miura J, et al . Role of the 84. Guasch G, Schober M, Pasolli HA, et al . Loss of TGFβ signaling
TAK1–NLK–STAT3 pathway in TGF-β-mediated mesoderm destabilizes homeostasis and promotes squamous cell carcinomas
induction. Genes Dev 2004; 18: 381–386. in stratified epithelia. Cancer Cell 2007; 12: 313–327.
66. Shim JH, Xiao C, Paschal AE, et al . TAK1, but not TAB 1 or 85. Bardeesy N, Cheng KH, Berger JH, et al . Smad4 is dispensable
TAB 2, plays an essential role in multiple signaling pathways for normal pancreas development yet critical in progression
in vivo. Genes Dev 2005; 19: 2668–2681. and tumor biology of pancreas cancer. Genes Dev 2006; 20:
67. Yamashita M, Fatyol K, Jin C, et al . TRAF6 mediates Smad- 3130–3146.
independent activation of JNK and p38 by TGF-β. Mol Cell 2008; 86. Takaku K, Oshima M, Miyoshi H, et al . Intestinal tumorigenesis
31: 918–924. in compound mutant mice of both Dpc4 (Smad4) and Apc genes.
68. Hutchison N, Hendry BM, Sharpe CC. Rho isoforms have dis- Cell 1998; 92: 645–656.
tinct and specific functions in the process of epithelial to mes- 87. Gomis RR, Alarcon C, He W, et al . A FoxO–Smad synexpres-
enchymal transition in renal proximal tubular cells. Cell Signal sion group in human keratinocytes. Proc Natl Acad Sci U S A
2009; 21: 1522–1531. 2006; 103: 12747–12752.
69. Bhowmick NA, Ghiassi M, Bakin A, et al . Transforming growth 88. Seoane J, Le HV, Shen L, et al . Integration of Smad and fork-
factor-β1 mediates epithelial to mesenchymal transdifferentiation
head pathways in the control of neuroepithelial and glioblastoma
through a RhoA-dependent mechanism. Mol Biol Cell 2001; 12:
cell proliferation. Cell 2004; 117: 211–223.
27–36.
89. Pardali K, Kurisaki A, Moren A, et al . Role of Smad pro-
70. Lee MK, Pardoux C, Hall MC, et al . TGF-β activates Erk MAP
teins and transcription factor Sp1 in p21(Waf1/Cip1) regula-
kinase signalling through direct phosphorylation of ShcA. EMBO
tion by transforming growth factor-β. J Biol Chem 2000; 275:
J 2007; 26: 3957–3967.
29244–29256.
71. Ozdamar B, Bose R, Barrios-Rodiles M, et al . Regulation of the
90. Datto MB, Li Y, Panus JF, et al . Transforming growth factor β
polarity protein Par6 by TGFβ receptors controls epithelial cell
induces the cyclin-dependent kinase inhibitor p21 through a p53-
plasticity. Science 2005; 307: 1603–1609.
independent mechanism. Proc Natl Acad Sci U S A 1995; 92:
72. Qiu T, Wu X, Zhang F, et al . TGF-β type II receptor phospho-
5545–5549.
rylates PTH receptor to integrate bone remodelling signalling.
91. Hannon GJ, Beach D. p15INK4B is a potential effector of TGF-
Nature Cell Biol 2010; 12: 224–234.
β-induced cell cycle arrest. Nature 1994; 371: 257–261.
73. Derynck R, Zhang YE. Smad-dependent and Smad-independent
92. Siegel PM, Massagué J. Cytostatic and apoptotic actions of TGF-
pathways in TGF-β family signalling. Nature 2003; 425:
β in homeostasis and cancer. Nature Rev Cancer 2003; 3:
577–584.
807–821.
74. Markowitz S, Wang J, Myeroff L, et al . Inactivation of the type
93. Scandura JM, Boccuni P, Massagué J, et al . Transforming
II TGF-β receptor in colon cancer cells with microsatellite
growth factor β-induced cell cycle arrest of human hematopoi-
instability. Science 1995; 268: 1336–1338.
etic cells requires p57KIP2 up-regulation. Proc Natl Acad Sci U
75. Grady WM, Myeroff LL, Swinler SE, et al . Mutational inactiva-
tion of transforming growth factor β receptor type II in microsatel- S A 2004; 101: 15231–15236.
lite stable colon cancers. Cancer Res 1999; 59: 320–324. 94. Staller P, Peukert K, Kiermaier A, et al . Repression of p15INK4b
76. Izumoto S, Arita N, Ohnishi T, et al . Microsatellite instability expression by Myc through association with Miz-1. Nature Cell
and mutated type II transforming growth factor-β receptor gene Biol 2001; 3: 392–399.
in gliomas. Cancer Lett 1997; 112: 251–256. 95. Seoane J, Le HV, Massague J. Myc suppression of the p21(Cip1)
77. Wang D, Kanuma T, Mizunuma H, et al . Analysis of specific Cdk inhibitor influences the outcome of the p53 response to DNA
gene mutations in the transforming growth factor-β signal trans- damage. Nature 2002; 419: 729–734.
duction pathway in human ovarian cancer. Cancer Res 2000; 60: 96. Sikder HA, Devlin MK, Dunlap S, et al . Id proteins in cell
4507–4512. growth and tumorigenesis. Cancer Cell 2003; 3: 525–530.
78. Goggins M, Shekher M, Turnacioglu K, et al . Genetic alterations 97. Azar R, Alard A, Susini C, et al . 4E-BP1 is a target of Smad4
of the transforming growth factor β receptor genes in pancreatic essential for TGFβ-mediated inhibition of cell proliferation.
and biliary adenocarcinomas. Cancer Res 1998; 58: 5329–5332. EMBO J 2009; 28: 3514–3522.
79. Chen T, Carter D, Garrigue-Antar L, et al . Transforming growth 98. Jang CW, Chen CH, Chen CC, et al . TGF-β induces apoptosis
factor β type I receptor kinase mutant associated with metastatic through Smad-mediated expression of DAP-kinase. Nature Cell
breast cancer. Cancer Res 1998; 58: 4805–4810. Biol 2002; 4: 51–58.
80. Forrester E, Chytil A, Bierie B, et al . Effect of conditional 99. Ohgushi M, Kuroki S, Fukamachi H, et al . Transforming growth
knockout of the type II TGF-β receptor gene in mammary epithe- factor β-dependent sequential activation of Smad, Bim, and
lia on mammary gland development and polyomavirus middle caspase-9 mediates physiological apoptosis in gastric epithelial
T antigen induced tumor formation and metastasis. Cancer Res cells. Mol Cell Biol 2005; 25: 10017–10028.
2005; 65: 2296–2302. 100. Yoo J, Ghiassi M, Jirmanova L, et al . Transforming growth
81. Lu SL, Herrington H, Reh D, et al . Loss of transforming growth factor-β-induced apoptosis is mediated by Smad-dependent
factor-β type II receptor promotes metastatic head-and-neck expression of GADD45b through p38 activation. J Biol Chem
squamous cell carcinoma. Genes Dev 2006; 20: 1331–1342. 2003; 278: 43001–43007.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
216 E Meulmeester and P ten Dijke

101. Kim SG, Jong HS, Kim TY, et al . Transforming growth factor- 122. Miettinen PJ, Ebner R, Lopez AR, et al . TGF-β induced trans-
β1 induces apoptosis through Fas ligand-independent activation differentiation of mammary epithelial cells to mesenchymal
of the Fas death pathway in human gastric SNU-620 carcinoma cells: involvement of type I receptors. J Cell Biol 1994; 127:
cells. Mol Biol Cell 2004; 15: 420–434. 2021–2036.
102. Sakakura T, Sakagami Y, Nishizuka Y. Accelerated mammary 123. Deckers M, van Dinther M, Buijs J, et al . The tumor suppressor
cancer development by fetal salivary mesenchyma isografted to Smad4 is required for transforming growth factor β-induced
adult mouse mammary epithelium. J Natl Cancer Inst 1981; 66: epithelial to mesenchymal transition and bone metastasis of breast
953–959. cancer cells. Cancer Res 2006; 66: 2202–2209.
103. Joseph H, Gorska AE, Sohn P, et al . Overexpression of a kinase- 124. Valcourt U, Kowanetz M, Niimi H, et al . TGF-β and the Smad
deficient transforming growth factor-β type II receptor in mouse signaling pathway support transcriptomic reprogramming during
mammary stroma results in increased epithelial branching. Mol epithelial–mesenchymal cell transition. Mol Biol Cell 2005; 16:
Biol Cell 1999; 10: 1221–1234. 1987–2002.
104. Bhowmick NA, Chytil A, Plieth D, et al . TGF-β signaling in 125. Dzwonek J, Preobrazhenska O, Cazzola S, et al . Smad3 is a key
fibroblasts modulates the oncogenic potential of adjacent epithelia. nonredundant mediator of transforming growth factor β signaling
Science 2004; 303: 848–851. in Nme mouse mammary epithelial cells. Mol Cancer Res 2009;
105. Cheng N, Bhowmick NA, Chytil A, et al . Loss of TGF-β type 7: 1342–1353.
II receptor in fibroblasts promotes mammary carcinoma growth 126. Piek E, Moustakas A, Kurisaki A, et al . TGF-(β) type I
and invasion through upregulation of TGF-α-, MSP- and HGF- receptor/ALK-5 and Smad proteins mediate epithelial to mes-
mediated signaling networks. Oncogene 2005; 24: 5053–5068. enchymal transdifferentiation in NMuMG breast epithelial cells.
106. Reimann M, Lee S, Loddenkemper C, et al . Tumor stroma- J Cell Sci 1999; 112: 4557–4568.
derived TGF-β limits myc-driven lymphomagenesis via Suv39h1- 127. Gregory PA, Bracken CP, Bert AG, et al . MicroRNAs as regu-
dependent senescence. Cancer Cell 2010; 17: 262–272. lators of epithelial–mesenchymal transition. Cell Cycle 2008; 7:
107. Myeroff LL, Parsons R, Kim SJ, et al . A transforming growth 3112–3118.
factor β receptor type II gene mutation common in colon 128. Proetzel G, Pawlowski SA, Wiles MV, et al . Transforming
and gastric but rare in endometrial cancers with microsatellite growth factor-β3 is required for secondary palate fusion. Nature
instability. Cancer Res 1995; 55: 5545–5547. Genet 1995; 11: 409–414.
108. Parsons R, Myeroff LL, Liu B, et al . Microsatellite instability 129. Kaartinen V, Voncken JW, Shuler C, et al . Abnormal lung devel-
and mutations of the transforming growth factor β type II receptor opment and cleft palate in mice lacking TGF-β3 indicates defects
gene in colorectal cancer. Cancer Res 1995; 55: 5548–5550. of epithelial–mesenchymal interaction. Nature Genet 1995; 11:
109. Schutte M, Hruban RH, Hedrick L, et al . DPC4 gene in various 415–421.
tumor types. Cancer Res 1996; 56: 2527–2530. 130. Sanford LP, Ormsby I, Gittenberger-de Groot AC, et al . TGFβ2
110. Hahn SA, Schutte M, Hoque AT, et al . DPC4, a candidate tumor knockout mice have multiple developmental defects that are non-
suppressor gene at human chromosome 18q21.1. Science 1996; overlapping with other TGFβ knockout phenotypes. Development
271: 350–353. 1997; 124: 2659–2670.
111. Vincent F, Hagiwara K, Ke Y, et al . Mutation analysis of the 131. Cui W, Fowlis DJ, Bryson S, et al . TGFβ1 inhibits the formation
transforming growth factor β type II receptor in sporadic human of benign skin tumors, but enhances progression to invasive
cancers of the pancreas, liver, and breast. Biochem Biophys Res spindle carcinomas in transgenic mice. Cell 1996; 86: 531–542.
Commun 1996; 223: 561–564. 132. Hoot KE, Lighthall J, Han G, et al . Keratinocyte-specific Smad2
112. Jones E, Pu H, Kyprianou N. Targeting TGF-β in prostate cancer: ablation results in increased epithelial–mesenchymal transition
therapeutic possibilities during tumor progression. Expert Opin during skin cancer formation and progression. J Clin Invest 2008;
Ther Targets 2009; 13: 227–234. 118: 2722–2732.
113. Jennings MT, Pietenpol JA. The role of transforming growth 133. Shipitsin M, Campbell LL, Argani P, et al . Molecular definition
factor β in glioma progression. J Neurooncol 1998; 36: 123–140. of breast tumor heterogeneity. Cancer Cell 2007; 11: 259–273.
114. Takenoshita S, Mogi A, Tani M, et al . Absence of mutations in 134. Mani SA, Guo W, Liao MJ, et al . The epithelial–mesenchymal
the analysis of coding sequences of the entire transforming growth transition generates cells with properties of stem cells. Cell 2008;
factor-β type II receptor gene in sporadic human breast cancers. 133: 704–715.
Oncol Rep 1998; 5: 367–371. 135. Ikushima H, Todo T, Ino Y, et al . Autocrine TGF-β signaling
115. Prall F. Tumour budding in colorectal carcinoma. Histopathology maintains tumorigenicity of glioma-initiating cells through Sry-
2007; 50: 151–162. related HMG-box factors. Cell Stem Cell 2009; 5: 504–514.
116. Kalluri R, Weinberg RA. The basics of epithelial–mesenchymal 136. Nguyen DX, Bos PD, Massagué J. Metastasis: from dissemina-
transition. J Clin Invest 2009; 119: 1420–1428. tion to organ-specific colonization. Nature Rev Cancer 2009; 9:
117. Tarin D, Thompson EW, Newgreen DF. The fallacy of epithelial 274–284.
mesenchymal transition in neoplasia. Cancer Res 2005; 65: 137. Bos PD, Nguyen DX, Massagué J. Modeling metastasis in the
5996–6000. mouse. Curr Opin Pharmacol 2010; 10: 571–577.
118. Moustakas A, Heldin CH. Signaling networks guiding epithe- 138. Kang Y, Siegel PM, Shu W, et al . A multigenic program medi-
lial–mesenchymal transitions during embryogenesis and cancer ating breast cancer metastasis to bone. Cancer Cell 2003; 3:
progression. Cancer Sci 2007; 98: 1512–1520. 537–549.
119. Xu J, Lamouille S, Derynck R. TGF-β-induced epithelial to mes- 139. Minn AJ, Gupta GP, Siegel PM, et al . Genes that mediate breast
enchymal transition. Cell Res 2009; 19: 156–172. cancer metastasis to lung. Nature 2005; 436: 518–524.
120. Berx G, Raspe E, Christofori G, et al . Pre-EMTing metastasis? 140. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and
Recapitulation of morphogenetic processes in cancer. Clin Exp soil’ hypothesis revisited. Nature Rev Cancer 2003; 3: 453–458.
Metastasis 2007; 24: 587–597. 141. Buck MB, Fritz P, Dippon J, et al . Prognostic significance of
121. Huber MA, Kraut N, Beug H. Molecular requirements for epithe- transforming growth factor β receptor II in estrogen receptor-
lial–mesenchymal transition during tumor progression. Curr Opin negative breast cancer patients. Clin Cancer Res 2004; 10:
Cell Biol 2005; 17: 548–558. 491–498.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
TGF-β in cancer 217

142. Biswas S, Guix M, Rinehart C, et al . Inhibition of TGF-β with 160. Castriconi R, Cantoni C, Della CM, et al . Transforming growth
neutralizing antibodies prevents radiation-induced acceleration factor β1 inhibits expression of NKp30 and NKG2D receptors:
of metastatic cancer progression. J Clin Invest 2007; 117: consequences for the NK-mediated killing of dendritic cells. Proc
1305–1313. Natl Acad Sci U S A 2003; 100: 4120–4125.
143. Muraoka RS, Koh Y, Roebuck LR, et al . Increased malignancy 161. Kopp HG, Placke T, Salih HR. Platelet-derived transforming
of Neu-induced mammary tumors overexpressing active trans- growth factor-β down-regulates NKG2D thereby inhibiting nat-
forming growth factor β1. Mol Cell Biol 2003; 23: 8691–8703. ural killer cell antitumor reactivity. Cancer Res 2009; 69:
144. Siegel PM, Shu W, Cardiff RD, et al . Transforming growth fac- 7775–7783.
tor β signaling impairs Neu-induced mammary tumorigenesis 162. Friese MA, Wischhusen J, Wick W, et al . RNA interference tar-
while promoting pulmonary metastasis. Proc Natl Acad Sci U S geting transforming growth factor-β enhances NKG2D-mediated
A 2003; 100: 8430–8435. antiglioma immune response, inhibits glioma cell migration and
145. Giampieri S, Manning C, Hooper S, et al . Localized and invasiveness, and abrogates tumorigenicity in vivo. Cancer Res
reversible TGFβ signalling switches breast cancer cells from cohe- 2004; 64: 7596–7603.
sive to single cell motility. Nature Cell Biol 2009; 11: 1287–1296. 163. Spearman M, Taylor WR, Greenberg AH, et al . Antisense oli-
146. Guise TA, Yin JJ, Taylor SD, et al . Evidence for a causal role godeoxyribonucleotide inhibition of TGF-β1 gene expression and
of parathyroid hormone-related protein in the pathogenesis of alterations in the growth and malignant properties of mouse
human breast cancer-mediated osteolysis. J Clin Invest 1996; 98: fibrosarcoma cells. Gene 1994; 149: 25–29.
1544–1549. 164. Wu RS, Kobie JJ, Besselsen DG, et al . Comparative analysis of
147. Yin JJ, Selander K, Chirgwin JM, et al . TGF-β signaling block- IFN-gamma B7.1 and antisense TGF-β gene transfer on the
ade inhibits PTHrP secretion by breast cancer cells and bone tumorigenicity of a poorly immunogenic metastatic mammary
metastases development. J Clin Invest 1999; 103: 197–206. carcinoma. Cancer Immunol Immunother 2001; 50: 229–240.
148. Kondo H, Guo J, Bringhurst FR. Cyclic adenosine monophos- 165. Hau P, Jachimczak P, Schlingensiepen R, et al . Inhibition of
phate/protein kinase A mediates parathyroid hormone/parathyroid TGF-β2 with AP 12009 in recurrent malignant gliomas: from
hormone-related protein receptor regulation of osteoclastogene- preclinical to phase I/II studies. Oligonucleotides 2007; 17:
sis and expression of RANKL and osteoprotegerin mRNAs by 201–212.
marrow stromal cells. J Bone Miner Res 2002; 17: 1667–1679. 166. Schlingensiepen KH, Fischer-Blass B, Schmaus S, et al . Anti-
149. Kingsley LA, Fournier PG, Chirgwin JM, et al . Molecular biol- sense therapeutics for tumor treatment: the TGF-β2 inhibitor AP
ogy of bone metastasis. Mol Cancer Ther 2007; 6: 2609–2617. 12009 in clinical development against malignant tumors. Recent
150. Kang Y, He W, Tulley S, et al . Breast cancer bone metastasis Results Cancer Res 2008; 177: 137–150.
mediated by the Smad tumor suppressor pathway. Proc Natl Acad 167. Muraoka RS, Dumont N, Ritter CA, et al . Blockade of TGF-β
Sci U S A 2005; 102: 13909–13914. inhibits mammary tumor cell viability, migration, and metastases.
151. Petersen M, Pardali E, van der Horst G, et al . Smad2 and Smad3 J Clin Invest 2002; 109: 1551–1559.
have opposing roles in breast cancer bone metastasis by dif- 168. Yang YA, Dukhanina O, Tang B, et al . Lifetime exposure to a
ferentially affecting tumor angiogenesis. Oncogene 2010; 29: soluble TGF-β antagonist protects mice against metastasis without
1351–1361. adverse side effects. J Clin Invest 2002; 109: 1607–1615.
152. Padua D, Zhang XH, Wang Q, et al . TGFβ primes breast tumors 169. Rowland-Goldsmith MA, Maruyama H, Matsuda K, et al . Sol-
for lung metastasis seeding through angiopoietin-like 4. Cell uble type II transforming growth factor-β receptor attenuates
2008; 133: 66–77. expression of metastasis-associated genes and suppresses pan-
153. Torre-Amione G, Beauchamp RD, Koeppen H, et al . A highly creatic cancer cell metastasis. Mol Cancer Ther 2002; 1:
immunogenic tumor transfected with a murine transforming 161–167.
growth factor type β1 cDNA escapes immune surveillance. Proc 170. Bandyopadhyay A, Zhu Y, Malik SN, et al . Extracellular domain
Natl Acad Sci U S A 1990; 87: 1486–1490. of TGFβ type III receptor inhibits angiogenesis and tumor growth
154. Gorelik L, Flavell RA. Immune-mediated eradication of tumors in human cancer cells. Oncogene 2002; 21: 3541–3551.
through the blockade of transforming growth factor-β signaling 171. Dasch JR, Pace DR, Waegell W, et al . Monoclonal antibodies
in T cells. Nature Med 2001; 7: 1118–1122. recognizing transforming growth factor-β. Bioactivity neutral-
155. Diener KR, Woods AE, Manavis J, et al . Transforming growth ization and transforming growth factor β2 affinity purification.
factor-β-mediated signaling in T lymphocytes impacts on prostate- J Immunol 1989; 142: 1536–1541.
specific immunity and early prostate tumor progression. Lab 172. Nam JS, Suchar AM, Kang MJ, et al . Bone sialoprotein mediates
Invest 2009; 89: 142–151. the tumor cell-targeted prometastatic activity of transforming
156. Thomas DA, Massagué J. TGF-β directly targets cytotoxic T cell growth factor β in a mouse model of breast cancer. Cancer Res
functions during tumor evasion of immune surveillance. Cancer 2006; 66: 6327–6335.
Cell 2005; 8: 369–380. 173. Inman GJ, Nicolas FJ, Callahan JF, et al . SB-431542 is a potent
157. Arteaga CL, Hurd SD, Winnier AR, et al . Anti-transforming and specific inhibitor of transforming growth factor-β superfamily
growth factor (TGF)-β antibodies inhibit breast cancer cell tumori- type I activin receptor-like kinase (ALK) receptors ALK4, ALK5,
genicity and increase mouse spleen natural killer cell activity. and ALK7. Mol Pharmacol 2002; 62: 65–74.
Implications for a possible role of tumor cell/host TGF-β interac- 174. Hjelmeland MD, Hjelmeland AB, Sathornsumetee S, et al . SB-
tions in human breast cancer progression. J Clin Invest 1993; 92: 431542, a small molecule transforming growth factor-β-receptor
2569–2576. antagonist, inhibits human glioma cell line proliferation and
158. Ghiringhelli F, Menard C, Terme M, et al . CD4+CD25+ regu- motility. Mol Cancer Ther 2004; 3: 737–745.
latory T cells inhibit natural killer cell functions in a transform- 175. Matsuyama S, Iwadate M, Kondo M, et al . SB-431542 and
ing growth factor-β-dependent manner. J Exp Med 2005; 202: Gleevec inhibit transforming growth factor-β-induced prolifer-
1075–1085. ation of human osteosarcoma cells. Cancer Res 2003; 63:
159. Lee JC, Lee KM, Kim DW, et al . Elevated TGF-β1 secretion and 7791–7798.
down-modulation of NKG2D underlies impaired NK cytotoxicity 176. Uhl M, Aulwurm S, Wischhusen J, et al . SD-208, a novel trans-
in cancer patients. J Immunol 2004; 172: 7335–7340. forming growth factor β receptor I kinase inhibitor, inhibits

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
218 E Meulmeester and P ten Dijke

growth and invasiveness and enhances immunogenicity of murine 182. Bueno L, de Alwis DP, Pitou C, et al . Semi-mechanistic mod-
and human glioma cells in vitro and in vivo. Cancer Res 2004; elling of the tumour growth inhibitory effects of LY2157299, a
64: 7954–7961. new type I receptor TGF-β kinase antagonist, in mice. Eur J Can-
177. Ge R, Rajeev V, Ray P, et al . Inhibition of growth and metastasis cer 2008; 44: 142–150.
of mouse mammary carcinoma by selective inhibitor of transform- 183. Biswas S, Criswell TL, Wang SE, et al . Inhibition of transform-
ing growth factor-β type I receptor kinase in vivo. Clin Cancer ing growth factor-β signaling in human cancer: targeting a tumor
Res 2006; 12: 4315–4330. suppressor network as a therapeutic strategy. Clin Cancer Res
178. Ehata S, Hanyu A, Fujime M, et al . Ki26894, a novel trans- 2006; 12: 4142–4146.
forming growth factor-β type I receptor kinase inhibitor, inhibits 184. Wrzesinski SH, Wan YY, Flavell RA. Transforming growth
in vitro invasion and in vivo bone metastasis of a human breast factor-β and the immune response: implications for anticancer
cancer cell line. Cancer Sci 2007; 98: 127–133. therapy. Clin Cancer Res 2007; 13: 5262–5270.
179. Shinto O, Yashiro M, Kawajiri H, et al . Inhibitory effect of a 185. Geiger T, Cox J, Ostasiewicz P, et al . Super-SILAC mix for
TGFβ receptor type-I inhibitor, Ki26894, on invasiveness of quantitative proteomics of human tumor tissue. Nature Methods
scirrhous gastric cancer cells. Br J Cancer 2010; 102: 844–851. 2010; 7: 383–385.
180. Melisi D, Ishiyama S, Sclabas GM, et al . LY2109761, a novel 186. Yu X, Schneiderhan-Marra N, Joos TO. Protein microarrays for
transforming growth factor β receptor type I and type II dual personalized medicine. Clin Chem 2010; 56: 376–387.
inhibitor, as a therapeutic approach to suppressing pancreatic 187. Alizadeh AA, Eisen MB, Davis RE, et al . Distinct types of
cancer metastasis. Mol Cancer Ther 2008; 7: 829–840. diffuse large B-cell lymphoma identified by gene expression
181. Zhang B, Halder SK, Zhang S, et al . Targeting transforming profiling. Nature 2000; 403: 503–511.
growth factor-β signaling in liver metastasis of colon cancer.
Cancer Lett 2009; 277: 114–120.

Copyright  2010 Pathological Society of Great Britain and Ireland. J Pathol 2011; 223: 205–218
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

Вам также может понравиться