Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

60 Current Molecular Medicine 2008, 8, 60-67

Hypoxic Regulation of Metastasis via Hypoxia-Inducible Factors

Eelke H. Gort1, Arjan J. Groot1, Elsken van der Wall2, Paul J. van Diest1 and Marc A. Vooijs*,1

1
Department of Pathology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
2
Division of Internal Medicine and Dermatology, University Medical Center Utrecht, 3508 GA Utrecht, The
Netherlands
Abstract: Metastases formation is a major factor in disease progression and accounts for the majority of can-
cer deaths. The molecular mechanisms controlling invasion, dissemination to blood or lymphatic systems and
spread of tumor cells to distant organs are still poorly understood. Recent observations indicate that the meta-
static phenotype may already be present during the angiogenic switch of tumors. Intratumoral hypoxia corre-
lates with poor prognosis and enhanced metastases formation. The Hypoxia Inducible Factors (HIFs) are key
molecules in the hypoxic response and play critical roles during tumor cell expansion by regulating energy me-
tabolism and the induction of angiogenesis. Increasing evidence implicates HIF function in metastatic cell
characteristics, like epithelial to mesenchymal transition, cell detachment, invasion and tumor cell seeding.
Here, we review the link between tumor cell hypoxia and the acquisition of metastatic behavior. We hypothe-
size that polyclonal tumor selection by hypoxia enhances metastatic capacity by transcriptional control of key
regulators of metastasis. This polyclonal hypoxic gene profile potentially develops into a metastatic profile, driv-
ing metastasis formation. The hypoxic gene profile in primary tumors may therefore provide a prognostic indi-
cator in clinical decision-making.
Keywords: Hypoxia, cancer, metastasis, HIF, EMT, angiogenesis, polyclonality.

INTRODUCTION HYPOXIA AND HIF


Outgrowing metastases are the major cause of hu- In mammalian cells, the HIF transcriptional complex
man cancer mortality. The acquisition of full metastatic is a key regulator of the local and systemic responses
capacity of cancer cells requires several sequential to hypoxia that occur during normal development and
rate-limiting steps: i. cancer cells must detach from the (patho-) physiological processes [5]. Intratumoral hy-
primary tumor, ii. invade into the surrounding stroma, poxia, defined roughly when intercapillary distances
iii. intravasate into the blood or lymphatic vessels, iv. exceed 140 micron, correlates with poor prognosis and
spread into the capillary bed of distant organs, and v. has become an important clinical parameter [6,7]. Can-
extravasate and colonize into the recipient organs [1]. It cer cells adapt to hypoxic environments by converting
is well established that tumor cell specific alterations to glycolytic energy metabolism, induction of angio-
(cell autonomous) are not sufficient to drive the metas- genesis and cellular survival programs, and this acute
tatic cascade but that continuous interaction with the cellular response to hypoxia is mediated by the HIF
microenvironment (non-cell autonomous) is needed to basic helix-loop-helix (bHLH) transcription factor family
permit tumor cell survival. One of crucial rate-limiting [8]. HIF is a heterodimeric protein complex composed
events during human tumor growth is encountered of a constitutively expressed bèta subunit, HIF-1 or
when tumors outgrow the pre-existing vasculature lead- aryl hydrocarbon receptor nuclear translocator (ARNT),
ing to hypoxia. Intratumoral hypoxia is a hallmark of and an oxygen-regulated alpha subunit both of which
solid cancer. Hypoxic tumors have a higher tendency to belong to the bHLH-PAS (Per, ARNT, SIM) protein
metastasize and are clinically characterized by therapy family [9]. Mammalian cells have three HIF isoforms.
resistance and a poor prognosis [2,3]. Tumor cell hy- HIF-1 and HIF-2 have been studied best and will be
poxia affects various key steps during the metastatic considered here. HIF stability is tightly regulated at
cascade and selects for metastatic variants [4]. Central the posttranslational level by oxygen concentration.
components in the cellular hypoxia response are the Under normal oxygen tension (normoxia), HIF is con-
Hypoxia-inducible Factors (HIFs). Here, we review how stitutively hydroxylated and acetylated at multiple
hypoxia directly affects gene-expression programs driv- proline and lysine residues in the Oxygen Dependent
ing metastatic spread. We summarize the increasing Degradation Domain (ODDD) and on asparagine resi-
evidence implicating HIFs in several key steps in the dues in the c-terminal transactivation domain (N832).
metastatic cancer cascade. Finally, we formulate a These modifications enhance binding of HIF to the
concept of how the early hypoxic microenvironment Von Hippel-Lindau (VHL) protein, the recognition com-
drives the selection of polyclonal metastatic cells con- ponent of an E3 ubiquitin ligase complex and regulate
tributing to more aggressive tumor behavior. assembly of HIF/- heterodimers with transcriptional
co-activators. Prolyl hydroxylation marks HIF for
*Address correspondence to this author at the Department of Pathol- ubiquitination and proteosomal degradation (for a re-
ogy, University Medical Center Utrecht, 3508 GA Utrecht, The Neth- view see [10]). Thus, under normoxia HIF protein is
erlands; Tel: +31-(0)88-7558316; Fax: +31 (0)30 2544990; E-mail:
m.vooijs@umcutrecht.nl continuously degraded. Under hypoxic conditions the

1566-5240/08 $55.00+.00 © 2008 Bentham Science Publishers Ltd.


Hypoxic Regulation of Metastasis via Hypoxia-Inducible Factors Current Molecular Medicine, 2008, Vol. 8, No. 1 61

activity of prolyl hydroxylases is attenuated since they


require ferrous-ions and O2 for their function, which
leads to HIF stabilization. After nuclear translocation,
HIF/ARNT heterodimers regulate transcription by
binding hypoxia responsive elements (HRE) [11].

ONCOGENIC REGULATION OF HIF


HIF protein stability and expression can also be in-
creased by tumor specific genetic alterations in onco-
genes or tumor suppressor genes. For example, in the
Von Hippel Lindau hereditary cancer syndrome the loss
of VHL leads to attenuated proteosomal degradation of
HIF and predisposes to a variety of highly vascular-
ized malignant and benign neoplasms [12,13]. Also in
sporadic cancers deregulation of oncogenes and tumor
suppressor genes affects HIF signaling. For instance,
the phosphatidylinositol-3 kinase (PI 3-kinase)/Akt
kinase pathway directly impacts on HIF stability and
HER-2/neu overexpression or the tuberous sclerosis
complex gene TSC-2 (Tuberin) loss induce HIF-1
stabilization via an mammalian target of rapamycin
(mTOR)-dependent pathway [5]. Furthermore, HIF-1
stabilization depends on the TP53 tumor suppressor
protein [14]. Other pathways implicated in HIF regula-
tions are the mitogen-activated protein kinase (MAPK),
nuclear factor-kappaB (NF-B) and Notch signaling
pathways [15-17].

CLINICAL SIGNIFICANCE OF HIF


Direct oxygen measurements into tumours has re-
vealed that areas of necrosis are surrounded by poor
oxygenated areas [18]. In those areas hypoxia-induced
nuclear HIF-1 protein can be readily detected by im-
Fig. (1). Peri-necrotic staining of HIF-1. A) haematoxilin-
munohistochemistry (Fig. (1)). Overexpression of HIF-
eosin staining and B) immunohistochemical staining for HIF-
1 is associated with human cancer progression in
1 in endometriod endometrial carcinoma. Note the nuclear
epithelial cancers of the head and neck, cervix, colon,
peri-necrotic staining of HIF-1 protein (black arrows). Ne-
breast and lung (for a review see [19]). In breast can- crotic area indicated by N. Magnification 40 x.
cer, HIF-1 expression is an independent predictor of
poor clinical outcome, with a greater predictive value
then other hypoxic markers like carbonic anhydrase IX this process is never complete and some epithelial
(CAIX) [2,20]. HIF-1 expression correlates with a characteristics remain [25]. A hallmark of EMT is the
therapy resistance and primary tumor HIF-1 overex- loss of E-Cadherin, an adhesion molecule frequently
pression has been shown to predict presence of bone lost in human cancer and causal in tumor formation
marrow metastases in breast cancer patients [20,21]. and metastasis [26,27]. Loss of E-Cadherin enhances
Moreover, HIF-1 is not only expressed in primary tu- cancer cell motility and resistance to anoikis [26,28].
mors, but also in metastatic lesions and HIF-1 ex- EMT is a crucial process during normal development of
pression predicts early relapse [22,3]. Gene expression multicellular organisms [29]. EMT is controlled by AKT-,
profiling has revealed a unique set of transcriptional Wnt-, Notch-, and Hedgehog-signaling pathways and
targets in hypoxic breast cancers [23]. These clinical can be induced by a number of growth factors, like
studies suggest an important role for HIF in disease transforming growth factor  (TGF-), epidermal growth
progression. We will now discuss the molecular evi- factor (EGF), hepatocyte growth factor (HGF) and fi-
dence supporting this hypothesis. broblast growth factors (FGF) [29]. Several transcrip-
tion factors critical for mesoderm formation, like SNAI1
CANCER CELL DETACHMENT AND EMT (SNAIL1), ZEB and certain bHLH factors, are known to
induce EMT through repression of E-Cadherin and in-
Cancer cell detachment is the initial event in meta- duction of mesenchymal gene expression [30]. Recent
stases formation of carcinomas and is marked by the findings implicate the HIF pathway in EMT induction,
loss of cell adhesion molecules and the acquisition of acting through these mesodermal fate genes. In VHL-
mesenchymal phenotypes [24]. This process is termed deficient renal cell carcinomas, both HIF-1 and HIF-
epithelial to mesenchymal transition (EMT) although 2 are involved in the downregulation of E-Cadherin
62 Current Molecular Medicine, 2008, Vol. 8, No. 1 Gort et al.

expression [31-33]. One of the EMT regulating factors proficient areas. This process is thought to occur inde-
regulated by HIF signaling is SNAI1, a zinc-finger tran- pendent of the angiogenic switch and has been termed
scription factor indispensable for mesoderm develop- the “invasive switch” [45]. Scatter factor or hepatocyte
ment [33,34,31]. In addition to SNAI1, other known rep- growth factor (HGF) is a pleiotropic cytokine which
ressors of E-Cadherin, like TCF3 (E12/E47), ZEB1 stimulates proliferation and invasion through its recep-
(yEF1/ ZFHX1A), and ZEB2 (SIP1/ ZFHX1B), are tor, the tyrosine kinase proto-oncogene c-Met [46]. c-
upregulated in VHL-deficient renal cell carcinomas Met is frequently overexpressed or mutated in human
[31,32]. Using genetic screens in C. elegans, we re- cancers [47]. Hypoxia in a HIF-dependent manner di-
cently identified ceTWIST (helix-loop-helix-8 or HLH-8) rectly promotes invasion by inducing c-Met transcrip-
as a critical downstream target in hypoxia sensing [35]. tion and sensitizing cells to HGF stimulation. c-Met is
TWIST1 is directly regulated by HIF-2 under hypoxia. required and sufficient for hypoxia-induced invasive
TWIST1, the Saethre-Chotzen syndrome susceptibility growth [45,48,49]. Thus, hypoxia unleashes a positive
gene, is a master regulator of mesodermal fate and a feedback loop between HIF and c-Met that fuels inva-
bHLH repressor that like SNAI1, binds E-box contain- sive growth. c-Met-induced motility is also enhanced by
ing promoters. In invasive lobular breast cancer, hypoxic modulation of cell surface integrin signaling.
TWIST1 contributes to metastasis by promoting EMT Integrins are adhesion receptors with bidirectional sig-
through down regulation of E-Cadherin and high levels naling between plasma membrane and ECM, that play
of TWIST1 correlates with high disease recurrence in essential roles in motility and migration [50]. MG-63
breast and ovarian cancer [36-38]. Whereas SNAI1- human osteosarcoma cells exposed to the hypoxia-
mediated repression of E-Cadherin indirectly leads to mimicking agent CoCl2 upregulate both integrin 5 (the
activation of mesenchymal markers such as Vimentin fibronectin receptor) and integrin 2 (the alpha chain of
[39,40], TWIST1 can directly activate N-Cadherin ex- the collagen receptor), resulting in increased adhesion
pression, a marker for EMT progression [41]. Thus both [51]. Upregulation of the integrin 51 dimer via HIF-1
SNAI1 and TWIST1 appear to orchestrate the Cadherin might explain HER-2/neu-dependent fibronectin liga-
switch during EMT. A central role for hypoxia and HIF- tion, resulting in increased survival of mammary ade-
dependent regulation EMT provides an attractive model nocarcinoma cells, since HER-2/neu can regulate HIF
in which hypoxia in early tumorigenesis triggers cancer expression under normoxic conditions [52,53]. Con-
cell detachment and invasion. versely, integrin-dependent signaling affects HIF activ-
ity itself via integrin-linked kinase (ILK)-mediated AKT
activation [54]. Fibroblasts lacking HIF-1 fail to
EXTRACELLULAR MATRIX MODULATION upregulate the GTPase RhoA, a core transducer of
AND INVASION integrin signaling critical in stress fiber formation, dur-
Strong evidence for a role of hypoxia and HIF as ing hypoxia [55]. Thus hypoxia and HIF enhance motil-
regulators of ECM degradation and tumor cell migration ity by acting at multiple levels to facilitate migration to-
comes from the analysis of Erler and collegues who wards nutrient and oxygen rich areas within oxygen-
identified the ECM crosslinking enzyme lysyl oxidase deprived tissues.
(LOX) as a direct HIF-1 target [42]. LOX is a copper-
dependent amine oxidase that plays a critical role in ACIDOSIS AND INVASION
the biogenesis of connective tissue matrices by Cancer cells starved for oxygen and nutrients switch
crosslinking elastin and collagen and has been shown to anaerobic glycolysis (the Pasteur effect) as an
to enhance tumor cell migration [43]. LOX expression alternative energy source in a process that is
in hypoxic cancers correlates with poor patient survival predominantly HIF dependent [56]. Anaerobic
and inhibition of LOX activity in a mouse model of meta- respiration produces lactic acid and adenosine
static breast cancer significantly reduced liver/bone triphosphate (ATP) from glucose, which results in
metastases [42]. Although the mechanism whereby acidosis. Acidosis is a general feature of human tumors
LOX increases metastatic behavior is unresolved it and has been shown to promote tumor cell invasion by
may directly affect motility by regulating FAK- destruction of adjacent non-cancerous tissue and ECM
dependent adhesion to collagen. These data warrant degradation [56]. Importantly, tumor-induced acidosis
the development of LOX inhibitors as anti-metastatic can negate the effect of anti-cancer therapeutics that
therapeutics. Interestingly, LOX homologues LOXL2 only function under physiological pH [57]. To prevent
and LOXL3 interact and cooperate with SNAI1 in the intracellular acidosis, HIF induces the transcription of
downregulation of E-cadherin in carcinoma progression carbonic anhydrase XI (CAIX) that leads to increased
[44]. Regulation of matrix metalloproteinases (MMPs), -
HCO3 uptake, thereby neutralizing the intracellular pH
urokinase type plasminogen activator (uPAR) and albeit at the expense of extracellular acidosis, which is
Cathepsin D by hypoxia may provide additional means +
further increased by the H ions expelled out of the cell
for hypoxic ECM modulation [5]. by proton pumps [8]. In addition, HIF-1 induces the
expression of the glycolytic enzyme phosphoglucose
CELL MOTILITY AND INVASION isomerase (PGI or autocrine motility factor (AMF)), a
major cell motility stimulating factor [58,59]. Hypoxia-
Hypoxic tumor cells adopt a multitude of strategies induced cell motility can be inhibited by PGI inhibition.
in addition to neoangiogenesis to overcome oxygen Clearly, the numerous metabolic changes induced by
deprivation by inducing cell motility to colonize oxygen hypoxia, in part dependent on HIF, can promote
Hypoxic Regulation of Metastasis via Hypoxia-Inducible Factors Current Molecular Medicine, 2008, Vol. 8, No. 1 63

pendent on HIF, can promote metastatic behavior. This geting both DLL4 and VEGF. The recently discovered
has led to the concept that combination therapies tar- link between HIF and Notch signaling might play an
geting both angiogenesis as well as intracellular (pH) important role in controlling this balance [15].
metabolism/regulation may enhance therapeutic effi-
ciency in cancer [8].
SURVIVAL
ANGIOGENESIS AND INTRAVASATION Cancer cells continuously enter the circulation yet
only few seed and produce deadly metastatic tumors.
The production of pro-angiogenic factors by hypoxic
Generally, loss of interaction with the environment trig-
(cancer) cells that induce endothelial cell proliferation
gers apoptosis (anoikis). Cancer cells can escape
and vessel sprouting is perhaps the best-studied func-
anoikis by repression of apoptosis. For instance, over-
tion of HIF. The “angiogenic switch” is a hallmark of
expression of the neurotrophin receptor TrkB protects
human solid cancer progression [60,61]. In 1971, it was
from cell death induced by cell-matrix dissociation [78].
already proposed that the capacity of tumor cells to TrkB expression can be up regulated directly by HIF-1,
induce angiogenesis promotes tumor cell progression
but not HIF-2, under hypoxic conditions [79]. Further-
and that anti-angiogenic therapy may improve clinical
more, TrkB can regulate VEGF expression via HIF-1
outcome [62]. Key players in this process are vascular
[80]. In addition, c-Met signaling confers resistance to
endothelial growth factors (VEGF), and VEGF recep-
anoikis and supports anchorage independence [81].
tors 1 (VEGFR1 or Flt-1), which are directly regulated
Furthermore, SNAI1/2 and TWIST1 act on anti-
by HIF [63,64]. Significantly, VEGF-A activation was apoptotic and prosurvival pathways by antagonizing
found to be crucial for tumor angiogenesis in a mouse
p53- and c-Myc-induced apoptosis, respectively
model for multistage pancreatic cancer [65]. The VEGF
[82,83]. Direct evidence for a role of hypoxia in resis-
gradient secreted by tumor cells induces sprouting of
tance to anoikis is lacking, but hypoxia does increase
endothelial cells, which express VEGFR1 and VEGFR2
metastatic potential by promoting cell survival in metas-
[66]. Loss of HIF-1 in endothelial cells (EC) impairs
tatic colonies [84,85].
hypoxia-induced angiogenesis via a VEGF-VEGFR
autocrine loop, showing that hypoxia also affects EC
function directly, perhaps by impaired VEGFR expres- SEEDING
sion [67]. In addition to VEGF, other angiogenic factors
are regulated by hypoxia, like angiopoietin-1, -2 and -4 Secondary tumor formation follows arrest and seed-
(ANGPT1, -2 and -4), placental growth factor, and ing. Cells arrest in narrow capillary beds of the organ
platelet-derived growth factor-B [68,69]. Altogether, vasculature which is an active process of cell-cell/cell-
hypoxia-induced angiogenesis is emerging as an im- matrix interaction, involving among others, the G-
portant mechanism controlling metastatic capacity by protein-coupled CXC chemokine receptor 4 (CXCR4)
recruitment of blood vessels and lymphatic circulation. and the CXCR4 ligand stromal cell-derived factor-1
This has already resulted in therapeutic strategies, us- (SDF-1 or CXCL12), and integrin-fibronectin interac-
ing anti-angiogenic agents like VEGF receptor blocking tion [1]. Expression of CXCR4 in lung-, breast-, ovar-
®
agents (e.g. Bevacizumab or Avastin ) [70]. At face ian-, renal-, and prostate cancer has been implicated in
value, anti-angiogenic treatment may also have tumor increasing metastatic potential and expression of SDF1
promoting effects by inducing hypoxia and selection of is found in target tissue of metastases [86]. Staller et al.
metastatic variants that use alternative strategies (i.e showed for the first time that CXCR4 expression is
migration) to overcome nutrient deficiency. Paradoxi- regulated by hypoxia in a HIF-2 dependent manner
cally pro-angiogenic treatment of tumors by activating [87]. In addition, SDF-1 is also upregulated in VHL defi-
the Notch-Delta signaling pathway may also provide cient cells, suggesting an autocrine pathway [88]. In
therapeutic benefit [71]. Delta-like 4 Notch ligand non-small cell lung cancer cells, activation of the epi-
(DLL4) signaling through Notch1 and Notch4 receptors dermal growth factor receptor also results in up regula-
is critical for embryonic vascular development [72,73]. tion of CXCR4, which was dependent on PI 3-
VEGF produced by both hypoxic tumor cells and endo- kinase/AKT/mTOR-mediated activation of HIF-1
thelial cells upregulates DLL4 and Notch signaling in [89,90]. In addition, HIF-mediated induction of SDF-1
normal and tumor endothelial cells [74,75]. Blocking in endothelial cells functions to recruit circulating
DLL4/Notch signaling using neutralizing antibodies CXCR4-positive tumor cells or endothelial progenitor
stimulates non-productive angiogenic sprouting and cells (EPC) to ischemic tissue [88,91]. This provides
branching in tumors induced by VEGF and leads to three strategies by which the CXCR4-SDF-1 interaction
increased tumor hypoxia and decreased tumor growth influences cancer cell seeding; i. hypoxic tumor cells
[76,77]. Thus, a delicate balance between VEGF and express CXCR4 and seed after SDF-1 recognition in
DLL4/Notch signaling is required for proper (tumor) distant non-hypoxic tissue, ii. hypoxic tumor cells ex-
angiogenesis. Disruption of this signaling cascade re- press SDF-1 which might recruit CXCR4 expressing
sults in a decrease of functional angiogenesis (VEGF) cells by chemotaxis, and iii. hypoxic cancer cells ex-
or increase in non-functional angiogenesis (DLL4). In- pressing both CXCR4 and SDF-1 establish an
terestingly, anti-DLL4 blocking strategies also proved autocrine signaling pathway, which results cancer cell
effective in tumors resistant to anti-VEGF treatment, autonomy. These are interesting findings and indicate
providing rationale for new combination therapies tar- that disruption of the CXCR4-SDF axis may be ex-
64 Current Molecular Medicine, 2008, Vol. 8, No. 1 Gort et al.

ploited to target hypoxic tumor seeding, using blocking tions within tumors suggesting that some form of coop-
antibodies [92]. erative interaction exists between subclones that de-
termines the frequency of metastases [94]. Support for
this has been obtained from the discovery that primary
POLYCLONAL SELECTION OF METASTA- breast cancers already harbor the metastasis (poor-
SIS BY HYPOXIA prognosis) gene expression signature, suggesting that
Apart from the effects of oxygen deprivation on ge- most primary tumor cells have the capacity to metasta-
nomic stability and environmental selection, resulting in size rather than evolve through the emergence of rare
progression of more aggressive malignancy, hypoxia metastatic variants [97,98]. Remarkably, this signature
induces a gene expression program, which facilitates seems applicable to epithelial tumors in general, sug-
metastatic spread (Fig. (2)). Molecules involved in an- gesting this is in part an intrinsic biological property of
giogenesis, cell detachment, cell motility, invasiveness cancer cell populations [99]. In retrospect it seems ob-
and chemotaxis are under direct control of the hypoxia- vious to propose that this polyclonality and heterogene-
regulated HIF pathway. The long held paradigm and ity is driven in part by the presence of cancer stem
working model that continuous selection of rare spon- cells. Importantly, a significant fraction of signature
taneous variants within cancers emerge and drive tu- genes are stromal-derived suggesting a critical role for
mor formation and progression is under scrutiny [93]. tumor-host microenvironment in driving tumor progres-
Fidler and colleagues were among the first to demon- sion and metastasis [100]. This is underscored by the
strate that B16 melanoma cells have an intrinsic cellu- analysis that tumor-derived stromal expression signa-
lar heterogeneity with respect to subpopulations of tures might provide equally strong clinical classifiers as
cancer cells with different metastatic capacity [94,95]. tumor cell signatures [101].
This has recently been validated by Minn et al. who We would like to extend the polyclonal selection
observed that pre-existing subpopulations of MDA-MB- working model by proposing that intratumoral hypoxia
231 breast cancer cells determine bone or lung metas- is a driving force behind the polyclonal expansion of
tasis [96]. Importantly, this polyclonal heterogeneity metastatic subpopulations within primary tumors (Fig.
stabilizes the metastatic capacity of clonal subpopula- (3)). This can explain both the clonality of secondary
tumors, as well as early on-set whole tumor population
malignancy, because gene signatures of multiple
clones add up to constitute the poor-prognosis gene-
signature. Intratumoral hypoxia is not only restricted to
necrotic areas but occurs throughout tumors and,
therefore affects the tumor population as a whole. We
have screened both breast and endometrial carcino-
mas that display high HIF levels for activating HIF mu-
tations but have found none to date (Horree et al., ac-
cepted) [102]. This is consistent with a model for poly-
clonal adaptation rather than monoclonal selection of
metastatic clones. Indeed, deletion of HIF-1 in a
mouse model for breast cancer demonstrates that HIF
is critical for tumor progression and metastasis but not
for tumor initiation [103]. Further, breast cancers with
bone marrow metastatic cells had higher HIF-1 ex-
pression on the RNA and protein level than those with-
out metastatic cells in the bone marrow [21]. Hypoxia
(through HIF) regulates a myriad of metastagenic
genes. The hypoxia-response signature predicts prog-
nosis independently and contributes to the poor-
prognosis gene signature in breast cancer
[2,23,104,105]. Identification of intratumoral hypoxia
might serve as a prognostic factor in clinical decision-
making.

CONCLUSION
In conclusion, the direct effect of hypoxia on cell
adhesion molecules and matrix degrading enzymes
provides a framework for testable hypotheses in mouse
models and cell systems. From a clinical point of view,
Fig. (2). Hypoxic regulation of genes involved in metastasis.
identification of the molecular networks regulating en-
Genes are divided by functional implication in metastasis.
hanced metastatic behavior provide important anti-
Furthermore, up or down regulation under hypoxia and in-
cancer targets that may aid in the diagnosis and man-
volvement of HIF-1 or -2 is indicated.
Hypoxic Regulation of Metastasis via Hypoxia-Inducible Factors Current Molecular Medicine, 2008, Vol. 8, No. 1 65

Fig. (3). The hypoxic gene profile versus the metastatic gene profile. On the left, the hypoxic gene profile is illustrated. The oxy-
gen gradient is indicated from red (normal oxygen levels) to blue (hypoxia). Cells that lie in the periphery of the blood supply
become hypoxic and start expressing genes involved in metastasis as indicated by different colors. On the right, a metastatic
primary tumor is depicted, containing cells that express genes involved in each sequential step in metastasis formation, thereby
constituting the metastatic gene profile. More blood vessels are present here because of the stimulatory effects of hypoxia on
angiogenesis. Cells expressing gene products functionally involved in the different steps of metastasis formation are depicted in
concordance with these function; i.e. angiogenesis, cell detachment, invasion and seeding. The hypoxic gene profile might de-
velop into metastatic profile, and therefore provide a predictive value in clinical decision-making.

agement of metastatic disease. Further elucidation of [10] Maxwell, P.H., Pugh, C.W. and Ratcliffe, P.J. (2001) Adv.
the hypoxia/HIF-mediated transcriptional networks will Exp. Med. Biol., 502, 365-76.
[11] Pugh, C.W., Tan, C.C., Jones, R.W. and Ratcliffe, P.J.
provide exciting insights into cancer biology and high- (1991) Proc. Natl. Acad. Sci. U S A, 88, 10553-7.
light diagnostic and therapeutic avenues to be explored [12] Maher, E.R. (2004) Curr. Mol. Med., 4, 833-42.
in the near future. [13] Kaelin, W.G., Jr. (2002) Nat. Rev. Cancer, 2, 673-82.
[14] Ravi, R., Mookerjee, B., Bhujwalla, Z.M., Sutter, C.H., Arte-
mov, D., Zeng, Q., Dillehay, L. E., Madan, A., Semenza, G.L.
REFERENCES and Bedi, A. (2000) Genes Dev., 14, 34-44.
[15] Gustafsson, M.V., Zheng, X., Pereira, T., Gradin, K., Jin, S.,
[1] Gupta, G.P. and Massague, J. (2006) Cell, 127, 679-95. Lundkvist, J., Ruas, J.L., Poellinger, L., Lendahl, U. and Bon-
[2] Bos, R., van der Groep, P., Greijer, A.E., Shvarts, A., Meijer, desson, M. (2005) Dev. Cell, 9, 617-28.
S., Pinedo, H.M., Semenza, G. L., van Diest, P.J. and van [16] Mazure, N.M., Brahimi-Horn, M.C. and Pouyssegur, J. (2003)
der Wall, E. (2003) Cancer, 97, 1573-81. Curr. Pharm. Des., 9, 531-41.
[3] Dales, J.P., Garcia, S., Meunier-Carpentier, S., Andrac- [17] Zhou, J., Schmid, T. and Brune, B. (2003) Mol. Biol. Cell, 14,
Meyer, L., Haddad, O., Lavaut, M.N., Allasia, C., Bonnier, P. 2216-25.
and Charpin, C. (2005) Int. J. Cancer, 116, 734-9. [18] Vaupel, P., Schlenger, K., Knoop, C. and Hockel, M. (1991)
[4] Vaupel, P., Mayer, A. and Hockel, M. (2004) Methods Enzy- Cancer Res., 51, 3316-22.
mol., 381, 335-54. [19] Vaupel, P. (2004) Oncologist, 9 Suppl 5, 10-7.
[5] Semenza, G.L. (2003) Nat. Rev. Cancer, 3, 721-32. [20] Trastour, C., Benizri, E., Ettore, F., Ramaioli, A., Chamorey,
[6] Movsas, B., Chapman, J.D., Greenberg, R.E., Hanlon, A.L., E., Pouyssegur, J. and Berra, E. (2007) Int. J. Cancer, 120,
Horwitz, E.M., Pinover, W.H., Stobbe, C. and Hanks, G.E. 1451-8.
(2000) Cancer, 89, 2018-24. [21] Woelfle, U., Cloos, J., Sauter, G., Riethdorf, L., Janicke, F.,
[7] Hockel, M., Schlenger, K., Hockel, S. and Vaupel, P. (1999) van Diest, P., Brakenhoff, R. and Pantel, K. (2003) Cancer
Cancer Res., 59, 4525-8. Res., 63, 5679-84.
[8] Pouyssegur, J., Dayan, F. and Mazure, N.M. (2006) Nature, [22] Zhong, H., De Marzo, A.M., Laughner, E., Lim, M., Hilton,
441, 437-43. D.A., Zagzag, D., Buechler, P., Isaacs, W.B., Semenza, G.L.
[9] Wang, G.L., Jiang, B.H., Rue, E.A. and Semenza, G.L. and Simons, J.W. (1999) Cancer Res., 59, 5830-5.
(1995) Proc. Natl. Acad. Sci. U S A, 92, 5510-4.
66 Current Molecular Medicine, 2008, Vol. 8, No. 1 Gort et al.

[23] Chi, J.T., Wang, Z., Nuyten, D.S., Rodriguez, E. H., Schaner, [50] Danen, E.H. and Yamada, K.M. (2001) J. Cell Physiol., 189,
M.E., Salim, A., Wang, Y., Kristensen, G. B., Helland, A., 1-13.
Borresen-Dale, A. L., Giaccia, A., Longaker, M. T., Hastie, T., [51] Indovina, P., Ferrante, A., Rainaldi, G. and Santini, M.T.
Yang, G. P., van de Vijver, M. J. and Brown, P. O. (2006) (2006) Cell Commun. Adhes., 13, 185-98.
PLoS Med., 3, e47. [52] Laughner, E., Taghavi, P., Chiles, K., Mahon, P.C. and Se-
[24] Christofori, G. (2006) Nature, 441, 444-50. menza, G.L. (2001) Mol. Cell Biol., 21, 3995-4004.
[25] Christiansen, J.J. and Rajasekaran, A.K. (2006) Cancer [53] Spangenberg, C., Lausch, E.U., Trost, T.M., Prawitt, D., May,
Res., 66, 8319-26. A., Keppler, R., Fees, S.A., Reutzel, D., Bell, C., Schmitt, S.,
[26] Derksen, P.W., Liu, X., Saridin, F., van der Gulden, H., Ze- Schiffer, I.B., Weber, A., Brenner, W., Hermes, M., Sahin, U.,
venhoven, J., Evers, B., van Beijnum, J.R., Griffioen, A. W., Tureci, O., Koelbl, H., Hengstler, J.G. and Zabel, B.U. (2006)
Vink, J., Krimpenfort, P., Peterse, J.L., Cardiff, R.D., Berns, Cancer Res., 66, 3715-25.
A. and Jonkers, J. (2006) Cancer Cell, 10, 437-49. [54] Tan, C., Cruet-Hennequart, S., Troussard, A., Fazli, L., Cos-
[27] Perl, A.K., Wilgenbus, P., Dahl, U., Semb, H. and Christofori, tello, P., Sutton, K., Wheeler, J., Gleave, M., Sanghera, J.
G. (1998) Nature, 392, 190-3. and Dedhar, S. (2004) Cancer Cell, 5, 79-90.
[28] Vleminckx, K., Vakaet, L., Jr., Mareel, M., Fiers, W. and van [55] Greijer, A.E., van der Groep, P., Kemming, D., Shvarts, A.,
Roy, F. (1991) Cell, 66, 107-19. Semenza, G.L., Meijer, G.A., van de Wiel, M.A., Belien, J.A.,
[29] Huber, M.A., Kraut, N. and Beug, H. (2005) Curr. Opin. Cell van Diest, P.J. and van der Wall, E. (2005) J. Pathol., 206,
Biol., 17, 548-58. 291-304.
[30] Peinado, H., Olmeda, D. and Cano, A. (2007) Nat. Rev. [56] Gatenby, R.A. and Gillies, R.J. (2004) Nat. Rev. Cancer, 4,
Cancer, 7, 415-28. 891-9.
[31] Esteban, M.A., Tran, M.G., Harten, S.K., Hill, P., Castellanos, [57] Mahoney, B.P., Raghunand, N., Baggett, B. and Gillies, R.J.
M.C., Chandra, A., Raval, R., O'Brien T.S. and Maxwell, P.H. (2003) Biochem. Pharmacol., 66, 1207-18.
(2006) Cancer Res., 66, 3567-75. [58] Funasaka, T., Yanagawa, T., Hogan, V. and Raz, A. (2005)
[32] Krishnamachary, B., Zagzag, D., Nagasawa, H., Rainey, K., FASEB J., 19, 1422-30.
Okuyama, H., Baek, J. H. and Semenza, G.L. (2006) Cancer [59] Niizeki, H., Kobayashi, M., Horiuchi, I., Akakura, N., Chen, J.,
Res., 66, 2725-31. Wang, J., Hamada, J. I., Seth, P., Katoh, H., Watanabe, H.,
[33] Evans, A.J., Russell, R.C., Roche, O., Burry, T.N., Fish, J.E., Raz, A. and Hosokawa, M. (2002) Br. J. Cancer, 86, 1914-9.
Chow, V.W., Kim, W.Y., Saravanan, A., Maynard, M.A., Ger- [60] Ferrara, N. (2002) Nat. Rev. Cancer, 2, 795-803.
vais, M.L., Sufan, R.I., Roberts, A.M., Wilson, L.A., Betten, [61] Carmeliet, P., Dor, Y., Herbert, J.M., Fukumura, D., Brussel-
M., Vandewalle, C., Berx, G., Marsden, P.A., Irwin, M.S., mans, K., Dewerchin, M., Neeman, M., Bono, F., Abra-
Teh, B.T., Jewett, M. A. and Ohh, M. (2007) Mol. Cell Biol., movitch, R., Maxwell, P., Koch, C.J., Ratcliffe, P., Moons, L.,
27, 157-69. Jain, R.K., Collen, D. and Keshert, E. (1998) Nature, 394,
[34] Imai, T., Horiuchi, A., Wang, C., Oka, K., Ohira, S., Nikaido, 485-90.
T. and Konishi, I. (2003) Am. J. Pathol., 163, 1437-47. [62] Folkman, J. (1971) N. Engl. J. Med., 285, 1182-6.
[35] Gort, E.H., van Haaften, G., Verlaan, I., Groot, A.J., Plasterk, [63] Forsythe, J.A., Jiang, B.H., Iyer, N.V., Agani, F., Leung,
R.H., Shvarts, A., Suijkerbuijk, K.P., van Laar, T., van der S.W., Koos, R.D. and Semenza, G. L. (1996) Mol. Cell Biol.,
Wall, E., Raman, V., van Diest, P.J., Tijsterman, M. and 16, 4604-13.
Vooijs, M. (2007) Oncogene, doi:10.1038/sj.onc.1210795. [64] Gerber, H.P., Condorelli, F., Park, J. and Ferrara, N. (1997)
[36] Hosono, S., Kajiyama, H., Terauchi, M., Shibata, K., Ino, K., J. Biol. Chem., 272, 23659-67.
Nawa, A. and Kikkawa, F. (2007) Br. J. Cancer, 96, 314-20. [65] Inoue, M., Hager, J.H., Ferrara, N., Gerber, H.P. and Hana-
[37] Martin, T.A., Goyal, A., Watkins, G. and Jiang, W.G. (2005) han, D. (2002) Cancer Cell, 1, 193-202.
Ann. Surg. Oncol., 12, 488-96. [66] Carmeliet, P. (2005) Oncology, 69 Suppl 3, 4-10.
[38] Yang, J., Mani, S.A., Donaher, J.L., Ramaswamy, S., Itzyk- [67] Tang, N., Wang, L., Esko, J., Giordano, F.J., Huang, Y.,
son, R.A., Come, C., Savagner, P., Gitelman, I., Richardson, Gerber, H.P., Ferrara, N. and Johnson, R.S. (2004) Cancer
A. and Weinberg, R.A. (2004) Cell, 117, 927-39. Cell, 6, 485-95.
[39] Cano, A., Perez-Moreno, M.A., Rodrigo, I., Locascio, A., [68] Kelly, B.D., Hackett, S.F., Hirota, K., Oshima, Y., Cai, Z.,
Blanco, M.J., del Barrio, M.G., Portillo, F. and Nieto, M.A. Berg-Dixon, S., Rowan, A., Yan, Z., Campochiaro, P.A. and
(2000) Nat. Cell Biol., 2, 76-83. Semenza, G.L. (2003) Circ. Res., 93, 1074-81.
[40] Batlle, E., Sancho, E., Franci, C., Dominguez, D., Monfar, M., [69] Yamakawa, M., Liu, L.X., Date, T., Belanger, A.J., Vincent,
Baulida, J. and Garcia De Herreros, A. (2000) Nat. Cell Biol., K.A., Akita, G.Y., Kuriyama, T., Cheng, S.H., Gregory, R.J.
2, 84-9. and Jiang, C. (2003) Circ. Res., 93, 664-73.
[41] Alexander, N.R., Tran, N.L., Rekapally, H., Summers, C.E., [70] Gordon, M.S., Margolin, K., Talpaz, M., Sledge, G.W., Jr.,
Glackin, C. and Heimark, R.L. (2006) Cancer Res., 66, 3365- Holmgren, E., Benjamin, R., Stalter, S., Shak, S. and Adel-
9. man, D. (2001) J. Clin. Oncol., 19, 843-50.
[42] Erler, J.T., Bennewith, K.L., Nicolau, M., Dornhofer, N., [71] Thurston, G., Noguera-Troise, I. and Yancopoulos, G.D.
Kong, C., Le, Q.T., Chi, J.T., Jeffrey, S.S. and Giaccia, A.J. (2007) Nat. Rev. Cancer, 7, 327-31.
(2006) Nature, 440, 1222-6. [72] Krebs, L.T., Shutter, J.R., Tanigaki, K., Honjo, T., Stark, K.L.
[43] Payne, S.L., Fogelgren, B., Hess, A.R., Seftor, E.A., Wiley, and Gridley, T. (2004) Genes Dev., 18, 2469-73.
E.L., Fong, S.F., Csiszar, K., Hendrix, M.J. and Kirschmann, [73] Gale, N.W., Dominguez, M.G., Noguera, I., Pan, L., Hughes,
D.A. (2005) Cancer Res, 65, 11429-36. V., Valenzuela, D.M., Murphy, A.J., Adams, N.C., Lin, H.C.,
[44] Peinado, H., Del Carmen Iglesias-de la Cruz, M., Olmeda, Holash, J., Thurston, G. and Yancopoulos, G.D. (2004) Proc.
D., Csiszar, K., Fong, K.S., Vega, S., Nieto, M.A., Cano, A. Natl. Acad. Sci. U S A, 101, 15949-54.
and Portillo, F. (2005) EMBO J., 24, 3446-58. [74] Diez, H., Fischer, A., Winkler, A., Hu, C.J., Hatzopoulos,
[45] Pennacchietti, S., Michieli, P., Galluzzo, M., Mazzone, M., A.K., Breier, G. and Gessler, M. (2007) Exp. Cell Res., 313,
Giordano, S. and Comoglio, P. M. (2003) Cancer Cell, 3, 1-9.
347-61. [75] Hellstrom, M., Phng, L.K., Hofmann, J.J., Wallgard, E., Coul-
[46] Furge, K.A., Zhang, Y.W. and Vande Woude, G.F. (2000) tas, L., Lindblom, P., Alva, J., Nilsson, A.K., Karlsson, L.,
Oncogene, 19, 5582-9. Gaiano, N., Yoon, K., Rossant, J., Iruela-Arispe, M.L., Kalen,
[47] Birchmeier, C., Birchmeier, W., Gherardi, E. and Vande M., Gerhardt, H. and Betsholtz, C. (2007) Nature, 445, 776-
Woude, G.F. (2003) Nat. Rev. Mol. Cell Biol., 4, 915-25. 80.
[48] Hara, S., Nakashiro, K.I., Klosek, S.K., Ishikawa, T., Shintani, [76] Noguera-Troise, I., Daly, C., Papadopoulos, N.J., Coetzee,
S. and Hamakawa, H. (2006) Oral Oncol., 42, 16-22. S., Boland, P., Gale, N.W., Lin, H.C., Yancopoulos, G.D. and
[49] Eckerich, C., Zapf, S., Fillbrandt, R., Loges, S., Westphal, M. Thurston, G. (2006) Nature, 444, 1032-7.
and Lamszus, K. (2007) Int. J. Cancer, 121, 276-83.
Hypoxic Regulation of Metastasis via Hypoxia-Inducible Factors Current Molecular Medicine, 2008, Vol. 8, No. 1 67

[77] Ridgway, J., Zhang, G., Wu, Y., Stawicki, S., Liang, W.C., [92] Gonzalo, J.A., Lloyd, C. M., Peled, A., Delaney, T., Coyle,
Chanthery, Y., Kowalski, J., Watts, R.J., Callahan, C., Kas- A.J. and Gutierrez-Ramos, J.C. (2000) J. Immunol., 165,
man, I., Singh, M., Chien, M., Tan, C., Hongo, J.A., de Sau- 499-508.
vage, F., Plowman, G. and Yan, M. (2006) Nature, 444, [93] Nowell, P.C. (1971) Hum. Pathol., 2, 347-8.
1083-7. [94] Fidler, I.J. and Kripke, M.L. (1977) Science, 197, 893-5.
[78] Douma, S., Van Laar, T., Zevenhoven, J., Meuwissen, R., [95] Poste, G., Doll, J. and Fidler, I.J. (1981) Proc. Natl. Acad.
Van Garderen, E. and Peeper, D. S. (2004) Nature, 430, Sci. U S A, 78, 6226-30.
1034-9. [96] Minn, A.J., Kang, Y., Serganova, I., Gupta, G.P., Giri, D.D.,
[79] Martens, L.K., Kirschner, K.M., Warnecke, C. and Scholz, H. Doubrovin, M., Ponomarev, V., Gerald, W.L., Blasberg, R.
(2007) J. Biol. Chem., 282, 14379-88. and Massague, J. (2005) J. Clin. Invest., 115, 44-55.
[80] Nakamura, K., Martin, K.C., Jackson, J.K., Beppu, K., Woo, [97] van 't Veer, L.J., Dai, H., van de Vijver, M.J., He, Y.D., Hart,
C.W. and Thiele, C.J. (2006) Cancer Res., 66, 4249-55. A.A., Mao, M., Peterse, H.L., van der Kooy, K., Marton, M.J.,
[81] Longati, P., Albero, D. and Comoglio, P.M. (1996) Cell Death Witteveen, A.T., Schreiber, G.J., Kerkhoven, R.M., Roberts,
Differ., 3, 23-8. C., Linsley, P.S., Bernards, R. and Friend, S.H. (2002) Natu-
[82] Maestro, R., Dei Tos, A.P., Hamamori, Y., Krasnokutsky, S., re, 415, 530-6.
Sartorelli, V., Kedes, L., Doglioni, C., Beach, D.H. and Han- [98] van de Vijver, M.J., He, Y.D., van't Veer, L.J., Dai, H., Hart,
non, G.J. (1999) Genes Dev., 13, 2207-17. A.A., Voskuil, D.W., Schreiber, G.J., Peterse, J.L., Roberts,
[83] Wu, W.S., Heinrichs, S., Xu, D., Garrison, S.P., Zambetti, C., Marton, M.J., Parrish, M., Atsma, D., Witteveen, A., Glas,
G.P., Adams, J.M. and Look, A. T. (2005) Cell, 123, 641-53. A., Delahaye, L., van der Velde, T., Bartelink, H., Rodenhuis,
[84] Zhang, L. and Hill, R.P. (2007) Cancer Res., 67, 7789-97. S., Rutgers, E.T., Friend, S.H. and Bernards, R. (2002) N.
[85] Greijer, A.E. and van der Wall, E. (2004) J. Clin. Pathol., 57, Engl. J. Med., 347, 1999-2009.
1009-14. [99] Ramaswamy, S., Ross, K.N., Lander, E.S. and Golub, T.R.
[86] Muller, A., Homey, B., Soto, H., Ge, N., Catron, D., Bu- (2003) Nat. Genet., 33, 49-54.
chanan, M.E., McClanahan, T., Murphy, E., Yuan, W., Wag- [100] Liotta, L.A. and Kohn, E.C. (2001) Nature, 411, 375-9.
ner, S. N., Barrera, J.L., Mohar, A., Verastegui, E. and Zlot- [101] Roepman, P., de Koning, E., van Leenen, D., de Weger,
nik, A. (2001) Nature, 410, 50-6. R.A., Kummer, J.A., Slootweg, P. J. and Holstege, F.C.
[87] Staller, P., Sulitkova, J., Lisztwan, J., Moch, H., Oakeley, E.J. (2006) Genome Biol., 7, R117.
and Krek, W. (2003) Nature, 425, 307-11. [102] Vleugel, M.M., Greijer, A.E., van der Wall, E. and van Diest,
[88] Zagzag, D., Krishnamachary, B., Yee, H., Okuyama, H., P.J. (2005) Cancer Genet. Cytogenet., 163, 168-72.
Chiriboga, L., Ali, M.A., Melamed, J. and Semenza, G.L. [103] Liao, D., Corle, C., Seagroves, T.N. and Johnson, R. S.
(2005) Cancer Res., 65, 6178-88. (2007) Cancer Res., 67, 563-72.
[89] Phillips, R.J., Mestas, J., Gharaee-Kermani, M., Burdick, [104] Nuyten, D.S. and van de Vijver, M.J. (2006) Breast Dis., 26,
M.D., Sica, A., Belperio, J.A., Keane, M.P. and Strieter, R.M. 149-56.
(2005) J. Biol. Chem., 280, 22473-81. [105] Vleugel, M.M., Greijer, A.E., Shvarts, A., van der Groep, P.,
[90] Liu, Y.L., Yu, J.M., Song, X.R., Wang, X.W., Xing, L.G. and van Berkel, M., Aarbodem, Y., van Tinteren, H., Harris, A.L.,
Gao, B.B. (2006) Cancer Biol. Ther., 5, 1320-6. van Diest, P.J. and van der Wall, E. (2005) J. Clin. Pathol.,
[91] Ceradini, D.J., Kulkarni, A.R., Callaghan, M.J., Tepper, O.M., 58, 172-7.
Bastidas, N., Kleinman, M. E., Capla, J.M., Galiano, R.D.,
Levine, J.P. and Gurtner, G.C. (2004) Nat. Med., 10, 858-64.

Received: October 29, 2007 Revised: January 07, 2008 Accepted: January 07, 2008

You might also like