Sei sulla pagina 1di 24

REVIEW ARTICLE

published: 31 January 2012


doi: 10.3389/fonc.2012.00006

Current knowledge on pancreatic cancer


Juan Iovanna 1 *, Maria Cecilia Mallmann 2 , Anthony Gonalves 3 , Olivier Turrini 4 and Jean-Charles Dagorn 1
1
INSERM U624, Stress Cellulaire, Parc Scientique et Technologique de Luminy, Marseille, France
2
Centre dInvestigation Clinique de Marseille, Marseille, France
3
Dpartement dOncologie Mdicale, Institut Paoli-Calmettes, Marseille, France
4
Dpartement de Chirurgie Oncologique, Institut Paoli-Calmettes, Marseille, France

Edited by: Pancreatic cancer is the fourth leading cause of cancer death with a median survival of
Eric Solary, University Paris-Sud,
6 months and a dismal 5-year survival rate of 35%. The development and progression of
France
pancreatic cancer are caused by the activation of oncogenes, the inactivation of tumor sup-
Reviewed by:
Agns Nol, University of Lige, pressor genes, and the deregulation of many signaling pathways. Therefore, the strategies
Belgium targeting these molecules as well as their downstream signaling could be promising for
Francisco X. Real, Centro Nacional de the prevention and treatment of pancreatic cancer. However, although targeted therapies
Investigaciones Oncolgicas, Spain
for pancreatic cancer have yielded encouraging results in vitro and in animal models, these
Jean-Yves Scoazec, Universit Lyon
1, France ndings have not been translated into improved outcomes in clinical trials.This failure is due
*Correspondence: to an incomplete understanding of the biology of pancreatic cancer and to the selection of
Juan Iovanna, INSERM U624, Stress poorly efcient or imperfectly targeted agents. In this review, we will critically present the
Cellulaire, 163 Avenue de Luminy, CP current knowledge regarding the molecular, biochemical, clinical, and therapeutic aspects
915, 13288 Marseille Cedex 9, France.
of pancreatic cancer.
e-mail: juan.iovanna@inserm.fr
Keywords: pancreas, oncogenes, suppressor genes, signaling, cancer, surgery, neoadjuvants, chemoprevention

INTRODUCTION design of new drugs and the molecular selection of existing drugs
Pancreatic cancer is the fourth leading cause of cancer death with for targeted therapy. In the following sections we will summarize
a median survival of 6 months and a dismal 5-year survival rate what is known regarding the precursor lesions of pancreatic can-
of 35% and this gure has remained relatively unchanged over cer, the molecular aspects of pancreatic cancer, and how some of
the past 25 years (Jemal et al., 2006). Even for patients diagnosed these molecular pathways could be exploited for the prevention
with local disease, the 5-year survival rate is only 15%. Known or treatment of pancreatic cancer. In addition, we will describe
risk factors for the disease include cigaret smoking, chronic and the current knowledge on other major factors of pancreatic can-
hereditary pancreatitis, late-onset diabetes mellitus, and familial cer development such as inammation, telomerases, angiogenesis,
cancer syndromes. The lethal nature of pancreatic cancer stems miRNA, epigenetics and genetics factors, and pancreatic cancer
from its propensity to rapidly disseminate to the lymphatic system stem cells, presently under study as possible therapeutic targets.
and distant organs. The presence of occult or clinical metastases at We will also present the therapeutic strategies currently available.
the time of diagnosis together with the lack of effective chemother- Finally, we will describe some promising chemopreventive agents
apies contributes to the high mortality in patients with pancreatic and some new anticancer drugs which are in development.
cancer. Pancreatic cancer is one of the most intrinsically drug-
resistant tumors and resistance to chemotherapeutic agents is a PANCREATIC CANCER PRECURSOR LESIONS
major cause of treatment failure in pancreatic cancer. Gemcitabine It is supposed, although not formally established, that like
is the standard chemotherapeutic drug for patients with advanced other epithelial cancers pancreatic cancers do not arise de novo
pancreatic cancer after a phase III trial in 1997 that demonstrated but undergo a stepwise progression through histologically well-
a modest survival advantage of this agent over 5-FU (median dened non-invasive precursor lesions, culminating in frank,
survival 5.65 versus 4.41 months, respectively), but surprisingly invasive neoplasia. Although putative precursor lesions of pan-
this treatment improved alleviation of disease-related symptoms creatic cancer were rst documented over a century ago, it was
(Burris et al., 1997). Very recently, a polychemotherapy regimen only at the end of the last century that several lines of evidence
combining 5-FU, irinotecan, and oxaliplatin (FOLFIRINOX) was began to associate invasive pancreatic cancer with these lesions.
shown to nearly double overall survival compared to gemcitabine, For example, autopsy studies conrmed that the prevalence of
at the expense of a manageable but increased toxicity, limiting what is now recognized as precursor lesions increased with age,
its use to good performance status patients. In addition, overall thus paralleling the frequency of invasive pancreatic cancer. Sim-
survival was less than 12 months (Conroy et al., 2011). Therefore, ilarly, most surgically resected pancreas harboring invasive cancer
there is a dire need for designing new and targeted therapeutic also showed the presence of non-invasive intra-ductular lesions
strategies that can overcome the drug-resistance and improve the in the surrounding parenchyma, suggesting an etiologic associa-
clinical outcome for patients diagnosed with pancreatic cancer. tion (Cubilla and Fitzgerald, 1976; Andea et al., 2003; Schwartz
For this purpose, the knowledge on the molecular aspects of pan- and Henson, 2007). Most importantly, careful molecular analyses
creatic cancer is very important and is likely to be helpful in the over the last 10 years have unequivocally demonstrated that these

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 1


Iovanna et al. Pancreatic cancer

precursor lesions share many of the underlying genetic alterations


observed in the inltrating cancer, underscoring their precursor
status (Brat et al., 1998; Brockie et al., 1998). By the late 1990s,
several terminologies were in use to describe these non-invasive
ductal lesions, leading to considerable difculties in comparing
inter-institutional studies. Fortunately, in 1999 emerged a consen-
sus nomenclature for precursor lesions of pancreatic cancer. The
pancreatic intraepithelial neoplasia (PanIN) scheme for classi-
fying these lesions, rst proposed by Klimstra and Longnecker,
has since become a gold standard at academic centers worldwide
(Hruban et al., 2001, 2004). Briey, PanINs are microscopic lesions
in the smaller (less than 5 mm) pancreatic ducts. PanINs can
be papillary or at, and are composed of columnar to cuboidal
cells with varying amounts of mucin. PanINs are classied into
four groups, PanIN-1A, PanIN-1B (low-grade PanINs), PanIN-2
(intermediate grade PanINs), and PanIN-3 (high-grade PanINs),
reecting a progressive increase in histologic grade culminating
in invasive neoplasia. PanIN lesions of the lowest grade can be
at (1A) or papillary (1B) but are characterized by absence of FIGURE 1 | Altered genes in pancreatic cancer. At the upper part, in red,
nuclear abnormalities and conserved nuclear polarity. PanIN-2 are shown the gene alterations with gain-of-function, and at the lower part,
in green, are showed the genes with lost-of-function, that promote
lesions are architecturally slightly more complex than PanIN-1
pre-cancerous lesions PanIN and IPMN, PDAC, and metastasis.
lesions and show more nuclear changes including loss of nuclear
polarity, nuclear crowding, variation in nuclear size (pleomor-
phism), nuclear hyperchromasia, and nuclear pseudostratica- in several signaling pathways, such as EGFR, Akt, NFB, CCKR,
tion. Mitoses are rarely seen. In contrast, PanIN-3 lesions, also Hedgehog, etc., and their molecular crosstalk also play impor-
referred to as carcinoma-in situ, demonstrate widespread loss tant roles in the molecular pathogenesis of pancreatic cancer (see
of polarity, nuclear abnormalities, and frequent mitoses. How- Figure 1).
ever, as a pre-invasive lesion, PanIN-3 is still contained within the
basement membrane. ACTIVATION OF ONCOGENES
If PanINs are the most common proposed precursor lesions for THE Ras ACTIVATED PATHWAY
pancreatic cancer, they are not the only ones. Intraductal papil- Oncogenes can be activated through different mechanisms includ-
lary mucinous neoplasms (IPMNs) are other precursor lesions, so ing point mutation and amplication. The activation of the ras
called because they typically present as radiologically detectable oncogene has been observed in more than 90% of pancreatic can-
cysts in the pancreas (Winter et al., 2006). IPMNs are mucin- cers (Almoguera et al., 1988). The ras gene family encodes three
producing epithelial neoplasms, which arise within the main pan- 21-kDa membrane-bound proteins involved in signal transduc-
creatic duct or one of its branches, and have often, although not tion and mediating pleiotropic effects including cell proliferation
always, a papillary architecture (Schmitz-Winnenthal et al., 2003; and migration. Activated ras is involved in growth factor-mediated
Hruban et al., 2004). By denition, IPMNs are found in the larger signal transduction pathways. About 8090% of pancreatic can-
pancreatic ducts. Those that involve the main pancreatic ducts are cers harbor point mutations at codons 12, 13, and 61 in K-ras
designatedmain duct type,while those that involve the secondary (Almoguera et al., 1988). This is the highest level of K-ras alter-
branches of the main pancreatic duct are designated branch duct ation found in any human tumor type. The point mutation leads
type (Hruban et al., 2004; Longnecker et al., 2005). Similar to to the generation of a constitutively active form of ras. The con-
PanINs, the cystic precursor lesions also demonstrate a multistep stitutively activated ras binds to GTP and gives uncontrolled
histological and genetic progression to invasive neoplasia. stimulation signals to downstream signaling cascades, promoting
uncontrolled cell growth. K-ras mutations in pancreatic cancer
MOLECULAR MECHANISMS INVOLVED IN THE typically develop during the early phase of carcinogenesis and
PATHOGENESIS OF PANCREATIC CANCER patients with mutated K-ras have a shorter survival than patients
Intensive investigation of molecular pathogenesis will help iden- with wild-type K-ras, suggesting that the mutation of K-ras par-
tifying useful molecules for diagnosis, treatment, and prognosis ticipates in the initiation and progression of pancreatic cancer.
of pancreatic cancer. In the last years, considerable research has In addition to point mutations, amplication of ras is also fre-
focused on identifying molecular events associated with pancreatic quently observed in pancreatic cancers, suggesting that activation
carcinogenesis and their correlation with the pathological status. of the ras oncogene is an important molecular event in pancreatic
Multiple subsets of genes were found to be activated or inactivated cancers.
during the development and progression of pancreatic cancer. The A peptide vaccine that aims to stimulate immunity against
activation of oncogenes and the inactivation of tumor suppressor cancer cells with mutant Ras proteins has been tested as an adju-
genes are in part responsible for the initiation and progression vant treatment in patients with pancreatic cancer (Toubaji et al.,
of pancreatic cancers. Moreover, the deregulation of molecules 2008). An extension to this research investigated the effects of

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 2
Iovanna et al. Pancreatic cancer

combination therapy with mutant Ras peptide plus granulocyte THE CYCLOOXYGENASE PATHWAY
macrophage colony-stimulating factor (Gjertsen et al., 2001). For The cyclooxygenase (COX) enzymes promote the formation of
Ras to function, it must undergo post-translational modication prostaglandins, leading to the induction of cell growth. There
so that it can attach to the cell membrane. One essential step are two isoforms of the COX enzyme. COX-1 is produced at
involves the addition of a 15-carbon isoprenoid chain, mediated a constant rate and the prostaglandins formed are involved in
by farnesyltransferase. The therapeutic use of tipifarnib, a farne- several normal physiologic events. COX-2, in contrast, is an
syltransferase inhibitor (FTI), in combination with gemcitabine inducible enzyme, absent from most normal tissues. However,
was disappointing in a phase III trial (Van Cutsem et al., 2004). its synthesis is stimulated in inammatory and carcinogenic
This nding could be partly explained by the fact that K-ras can be processes by cytokines, growth factors, and other cancer pro-
alternatively prenylated by the addition of a 20-carbon isoprenoid moters. COX-2 has been shown to be increased in a variety of
moiety mediated by the enzyme geranylgeranyltransferase. More- cancers including pancreatic cancers. Several pancreatic cancer
over, FTIs work largely by inhibition of the cell cycle, but gem- cell lines strongly express COX-2. Immunohistochemical stud-
citabine needs cell cycle progression to be effective. To this end, ies have shown that about 50% of human pancreatic cancers
a dual inhibitor of farnesyltransferase and geranylgeranyltrans- over-express COX-2 and that COX-2 mRNA expression is much
ferase (L-778,123) was tested in a phase I trial in combination with higher in tumors than in normal surrounding tissue (Okami
radiotherapy for locally advanced pancreatic cancer (Martin et al., et al., 1999). Moreover, there is a positive association between
2004). Inhibition of farnesylation and sensitivity to radiotherapy ras mutation and COX-2 level because activated ras increases
was demonstrated in a patient-derived cell line. Further develop- the stability of COX-2 mRNA. Therefore, COX-2 appears to
ment of this drug was, nevertheless, halted owing to adverse cardiac be of signicance in pancreatic carcinogenesis and a crosstalk
effects. Other compounds that are in early phases of clinical test- between ras, NFB, Notch, and COX-2 in cellular signaling
ing after yielding promising laboratory results include romidepsin, might contribute to the molecular pathogenesis of pancreatic
a histone deacetylase inhibitor that inhibits Ras-mediated signal cancer.
transduction and thus causes cell cycle arrest, and farnesylthios- As indicated above, COX-2 and its metabolic product (PGE2)
alicylic acid (salirasib), which disrupts Ras from its membrane- play important roles in pancreatic cancer, suggesting that target-
binding site. These compounds seem to have clinical activity in ing COX-2 could provide a therapeutic benet. Several COX-2
combination with gemcitabine and further studies are necessary. inhibitors have shown activity in reducing tumor growth with dif-
MAP2K, the principal downstream component of Ras signaling, ferent mechanisms. Indomethacin, one of the COX-2 inhibitors,
has also been the subject of targeted inhibition. In a phase II trial, inhibits both isoforms of the COX enzyme while newer agents,
the inhibitor CI-1040 (PD184532) did not demonstrate enough such as celecoxib, inhibit only COX-2 and are more desirable for
anti-tumor activity to justify further development (Rinehart et al., clinical use. In an orthotopic pancreatic cancer animal model, cele-
2004). Nevertheless, combined inhibition of MAP2K and other coxib treatment showed inhibition of tumor growth, angiogenesis,
kinases (such as EGFR) has been effective in preclinical studies, and metastasis (Wei et al., 2004). Non-steroidal anti-inammatory
which suggests that this approach might still have a role in ther- drugs (NSAIDs) also show their inhibitory effect on COX-2 and
apy for pancreatic cancer (Jimeno et al., 2007; Takayama et al., PGE2, leading to a reduced incidence of tumor formation and a
2008). reduced number of tumors per animal. Experiments using two
NSAIDs, sulindac and NS398, in pancreatic cancer cell lines, have
THE NOTCH PATHWAY shown that both agents cause a dose-dependent inhibition of can-
Notch has also been considered as an oncogene involved in the cer cell growth. A combination of the agents, celecoxib and Zyo (a
pathogenesis of pancreatic cancer (Miyamoto et al., 2003). So 5-lipoxygenase inhibitor), has shown a reduction in the incidence
far, four Notch genes have been identied (Notch-1, Notch- and size of pancreatic tumors, and a reduced number of liver
2, Notch-3, and Notch-4) and ve Notch ligands (Dll-1, Dll- metastases in animal model. It has been reported that celecoxib
3, Dll-4, Jagged-1, and Jagged-2) have been found in mam- potentiates gemcitabine-induced growth inhibition through the
mals. Notch protein can be activated by interacting with its down-regulation of NFB activation and induction of apoptosis
ligands. Upon activation, Notch protein is cleaved by the - in pancreatic cancer cells (El-Rayes et al., 2004). In a clinical trial,
secretase, releasing intracellular Notch which translocates into patients with advanced pancreatic adenocarcinoma were given
the nucleus. The intracellular Notch associates with transcrip- celecoxib in combination with 5-uorouracil. Such association
tional factors which regulate the expression of target genes and was capable of inducing durable and objective responses, even
thus play an important role in both organ development and in gemcitabine-resistant pancreatic cancer (Milella et al., 2004).
pancreatic carcinogenesis. In fact, Notch signaling is frequently These results collectively suggest that COX-2 inhibition with other
deregulated in human pancreatic cancers (Buchler et al., 2005; novel agents could be useful in future clinical trials for pancreatic
Wang et al., 2006c,e). Notch signaling occurs downstream of Ras, cancer.
EGFR, and TGF- signaling in pancreatic tumorigenesis and pro-
motes tumor vascularization. Down-regulation of Notch-1 with THE HEPATOCYTE GROWTH FACTOR RECEPTOR PATHWAY
siRNA or curcumin (owing to the crosstalk between Notch and The MET oncogene, which encodes the receptor for hepato-
NFB signaling pathways) can inhibit cell growth and induce cyte growth factor (HGF), is overexpressed in 78% of pancreatic
apoptosis in pancreatic cancer cell lines in vitro (Wang et al., cancers (Furukawa et al., 1995). HGF is normally produced by
2006a,d). mesenchymal cells and acts on epithelial cells to promote tissue

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 3


Iovanna et al. Pancreatic cancer

regeneration. In hypoxic conditions, however, tumor-associated THE Wnt PATHWAY


broblasts produce HGF, which stimulates angiogenesis and Wnt signaling is involved in normal embryonic development and
tumor growth, enhances cell motility and extracellular matrix homeostatic self-renewal of a number of adult tissues. Three Wnt
breakdown and leads to invasion and metastasis. Targeting the signaling cascades have been described, the canonical Wnt-
HGF pathway with use of a synthetic competitive antagonist of catenin, the planar-cell polarity, and the WntCa2+ pathways.
HGF (Tomioka et al., 2001) and an antibody against the MET The former is the best known and has been implicated in a vari-
receptor (Jin et al., 2008) has yielded encouraging results in the ety of cancers including liver, colorectal, breast, prostate, renal,
laboratory setting. ARQ 197 is a MET receptor tyrosine kinase and hematological malignancies. Normally, -catenin is phospho-
inhibitor that is currently being tested in a phase II trial. A phase I rylated and targeted for degradation. However, binding of Wnt
study showed that it was well-tolerated by patients (Munshi et al., proteins results in activation of intracellular pathways that cause
2010). -catenin to enter the nucleus, where its interaction with the T-
cell factor (TCF) and lymphoid enhancer factor (LEF) families of
THE INSULIN-LIKE GROWTH FACTOR PATHWAY transcription factors leads to targeted gene expression. Any gain-
The insulin-like growth factor I (IGF-I) receptor, a transmembrane of-function mutation of activators or loss-of-function mutation
tyrosine kinase receptor, is overexpressed in 64% of pancreatic of inhibitors of Wnt signaling could lead to aberrant activation
cancers (Hakam et al., 2003). The IGF-I receptor has antiapop- of these signaling pathways, which could result in carcinogenesis
totic and growth-promoting effects and acts via multiple signal- and tumor progression. Aberrant activation occurs in 65% of pan-
ing cascades, including the PI3-Akt, MAPK, and STAT pathways. creatic cancers (Zeng et al., 2006). Inhibition of Wnt signaling to
Inhibition of the IGF-I receptor by the tyrosine kinase inhibitor reduce proliferation and increase apoptosis of pancreatic cancer
NVP-AEW541, a dominant-negative mutant, and RNA interfer- cells has been achieved in the laboratory setting by a variety of
ence have all been shown to reduce the growth of pancreatic cancer methods, including the use of -catenin-interacting protein 1, a
cells in vitro and in vivo, and increase chemotherapy-induced or dominant-negative mutant of LEF-1, and siRNA against -catenin
radiation-induced apoptosis (Piao et al., 2008). Concomitant inhi- or extracellular sulfatases (Nawroth et al., 2007; Pasca di Magliano
bition of K-ras increases the therapeutic effect of IGF-I receptor et al., 2007). Wnt signaling is positively regulated by the hedge-
antisense oligonucleotide (Shen et al., 2008). Human anti-IGF-I hog and SMAD4 signaling pathways (Romero et al., 2008), which
receptor antibodies have been reported to enhance the anti-tumor could be targets for a combined inhibitory therapeutic strategy.
effects of gemcitabine and EGFR inhibition in vivo (Beltran et al.,
2009).
THE CHEMOKINE RECEPTOR 4 AND ITS LIGAND SDF-1
The chemokine receptor 4 (CXCR4) and its ligand, SDF-1 have a
THE FOCAL ADHESION KINASE PATHWAY
role in tumor growth, angiogenesis, and particularly in metasta-
Focal adhesion kinase (FADK) is a cytoplasmic non-receptor tyro-
tic spread. In vitro blockade of CXCR4 inhibits pancreatic can-
sine kinase that mediates functions involved in cell motility and
cer growth through inhibition of the canonical Wnt pathway
survival and is closely related to the integrin signaling pathway.
(Wang et al., 2008). Furthermore, plerixafor, an antagonist of
Close to 80% of pancreatic cancers (Furuyama et al., 2006) express
CXCR4, reduces metastasis from pancreatic cancer cells positive
FADK and, importantly, it shares a pathway with the IGF-I receptor
for the CXCR4 and CD133 markers (the latter being a marker of
(Liu et al., 2008). The dual IGF-I receptorFADK inhibitor NVP-
pancreatic cancer stem cells) in vivo (Hermann et al., 2007).
TAE226 has shown signicant tumor-suppressive activity in vivo
(Liu et al., 2007).
OTHER ONCOGENES
THE Src PATHWAY Amplication of other oncogenes also plays a very important
Src is one of the nine members of the Src family of non-receptor role in the development and progression of pancreatic cancer.
protein tyrosine kinases. In normal conditions, Src is maintained The Akt-2 gene is amplied in 15% of pancreatic cancers while
in a phosphorylated and inactive form but it is activated in a Myb gene is amplied in 10% of pancreatic cancers. The ampli-
number of malignancies, including in 70% of pancreatic cancers cation of the oncogenes contributes to the stimulation of cell
(Hakam et al., 2003). Src has diverse roles in cell proliferation, growth and the progression of pancreatic cancers. There are exper-
survival, motility, invasiveness, resistance to chemotherapy, and imental evidences of up-regulation of other oncogenes including
angiogenesis. This protein acts via multiple signaling pathways Bcl-6, S100P, and Cyclin D1 (Mimeault et al., 2005). Up-regulation
and, therefore, is an ideal target for therapeutic intervention. Src of cyclin D1 has been found in pancreatic cancers and overex-
kinase inhibitors have been effective in suppressing pancreatic pression of cyclin D1 is associated with poor prognosis. Inhibit-
tumor growth and metastasis in vivo (Baker et al., 2006; Trevino ing cyclin D1 in pancreatic cancer cell lines leads to growth
et al., 2006; Ischenko et al., 2007). Dasatinib is an orally active inhibition and loss of tumorigenicity in nude mice (Kornmann
multitargeted kinase inhibitor of Src, BCRABL, platelet-derived et al., 1998). Recent studies have shown that overexpression of
growth factor receptor (PDGFR), ephrin type A receptor 2, and cyclin D1 promotes tumor cell growth and confers resistance
SCFR, and is licensed for the treatment of chronic myelogenous to cisplatin-mediated apoptosis in an elastasemyc transgene-
leukemias and acute lymphoblastic leukemias. Dasatinib is being expressing pancreatic tumor cell line, suggesting that cyclin D1
examined in a phase II trial in patients with metastatic pancreatic inuences the progression of pancreatic cancer (Biliran et al.,
cancer (Morton et al., 2010). 2005).

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 4
Iovanna et al. Pancreatic cancer

INACTIVATION OF TUMOR SUPPRESSOR GENES II TGF- receptors forms a heteromeric complex that triggers the
Inactivation of tumor suppressor genes is another important event phosphorylation of cytoplasmic SMAD2 and SMAD3. In turn,
for the initiation of pancreatic cancer. Tumor suppressor genes SMAD proteins form a complex with SMAD4, which translocates
can be activated by mutation, deletion, or hypermethylation. The into the nucleus to activate gene transcription. TGF- can also sig-
tumor suppressor genes targeted in pancreatic cancer include p16, nal via SMAD-independent pathways that involve Ras, PI3K, and
p53, SMAD4, and PTEN (see Figure 1). MAPK. TGF- mediates a wide range of physiological processes,
such as embryonic development, tissue repair, angiogenesis, and
THE p16/INK4a PATHWAY immunosuppression. TGF- also has a complex role in tumorige-
It has been shown that p16/INK4a inhibits the activity of cyclin D nesis, as it is tumor-suppressive in epithelial cells, but promotes
and of the CDK4/6 complex. CDK4 and CDK6 normally interact invasion and metastasis during the late stages of cancer progres-
with cyclin D to phosphorylate the retinoblastoma (Rb) pro- sion. Mutations of the TGFBR1 and TGFBR2 genes are found in
tein. The phosphorylation of Rb allows its dissociation from about 1 and 4% of pancreatic cancers, respectively (Goggins et al.,
a complex formed with the elongation factor 2 (E2F), allow- 1998b).
ing E2F to activate genes required for DNA synthesis along the The inactivation of DPC4 (Deleted in Pancreatic Cancer locus
cell cycle. p16/INK4a controls cell cycle progression through 4, Smad4) tumor suppressor gene is another common genetic
G1/S transition by inhibiting cyclin D and CDK4/6 mediated alteration identied in pancreatic cancer. The DPC4 gene encodes
phosphorylation of Rb, and therefore inhibiting cell growth. a 64-kDa protein, Smad 4, which plays roles in the inhibition of
Approximately 95% of pancreatic cancer patients have inacti- cell growth and angiogenesis. The inactivation of DPC4 is rela-
vated p16/INK4a in their tumors (about 40% by deletion, 40% tively specic to pancreatic cancer although it occurs with low
by mutation, and 15% by hypermethylation of its promoter; incidence in other cancers. It has been found that the DPC4
Schutte et al., 1997). Experimental studies have demonstrated tumor suppressor gene is deleted in approximately 50% of pan-
that transfection of wild-type p16/INK4a into human pancreatic creatic cancers (Cowgill and Muscarella, 2003). Up to 90% of
cancer cells results in decreased tumor cell proliferation in vitro pancreatic adenocarcinomas could harbor loss of heterozygosity.
and in vivo. Moreover, in pancreatic cancer patients the tumor DPC4 alterations occur relatively late in pancreatic carcinogen-
size is signicantly larger and the survival time is signicantly esis. The frequency of loss of DPC4 expression is signicantly
shorter when the p16/INK4a mutation is present, compared to higher in poorly differentiated pancreatic adenocarcinoma and
patients with wild-type p16/INK4a. These evidences suggest that the pancreatic cancer patients with intact DPC4 gene have signif-
p16/INK4a alterations participate in the aggressiveness of pancre- icantly longer survival after resection compared to patients with
atic cancer through its interaction with various cellular signaling mutant DPC4 gene. Furthermore, DPC4 inactivation is always
pathways. accompanied by inactivation of p16/INK4a, supporting its impor-
THE p53 PATHWAY tance in the pathogenesis of pancreatic cancer. It is noteworthy
In addition to p16/INK4a, another tumor suppressor gene, named that inactivation of SMAD4 abolishes TGF--mediated tumor-
p53, is well known to be involved in the control of cell cycle. suppressive functions while it maintains some tumor-promoting
p53 Binds to the p21/WAF1 promoter and stimulates the pro- TGF- responses, such as epithelialmesenchymal transition,
duction of p21/WAF1, which negatively regulates the complex which makes cells more invasive and more prone to migrate (Levy
consisting of cyclin D1 and CDK2, thereby arresting the cell at the and Hill, 2005).
G1 phase and inhibiting cell growth. The p53 also plays impor- TGF--based therapeutic strategies are currently in develop-
tant roles in the induction of apoptotic cell death. Inactivation of ment, including inhibitors of TGFBR1 and TGFBR2. AP 12009,
p53 during carcinogenesis can lead to uncontrolled cell growth an antisense oligonucleotide specic to TGF-2, is currently being
and increased cell survival. The p53 gene is inactivated in about tested in a phase III study of malignant melanoma, pancreatic
50% of pancreatic cancers through gene mutation and deletion. cancer, and colorectal carcinomas (Bonafoux and Lee, 2009).
It had been shown that p53 mutation results in alteration of the
3D structure of the p53 protein (Li et al., 1998), associated with OTHER TUMOR SUPPRESSOR GENES
shorter survival in patients with pancreatic cancer. In addition, As indicated earlier, p21/WAF1 is an inhibitor of CDK. It forms
alterations in the p53 gene are associated with K-ras mutations, complexes with cyclinA/CDK2 or cyclinD1/CDK4 and inhibits
suggesting a crosstalk and cooperative activity between p53 and their activity, causing cell cycle arrest in G1 phase. Loss of
K-ras in the molecular pathogenesis of pancreatic cancers. More- p21WAF1 activity has been observed in approximately 50% of
over, the status of p53 is important in mediating the specicity pancreatic cancers (Garcea et al., 2005). p27/CIP1 is another CDK
of chemotherapeutic agents on cancers. Loss of p53 function inhibitor which regulates cell cycle progression from G1 to S
could result in decreased sensitivity to certain types of chemother- phases. The loss of p27/CIP1 expression has also been observed in
apeutic agents. Therefore, the status of p53 could be a useful pancreatic cancers (Garcea et al., 2005). Another tumor suppres-
guide for those patients who are likely to respond to adjuvant sor gene, BRCA2, has been found to participate in DNA damage
chemotherapy. repair and mutations in BRCA2 have been linked to a signicantly
increased risk of pancreatic cancer. These evidences demonstrate
THE TGF- AND SMAD4 PATHWAY that inactivation of p21/WAF1, p27/CIP1, and BRCA2 tumor
TGF- is a cytokine secreted by epithelial, endothelial, hematopoi- suppressor genes is involved in the pathogenesis of pancreatic
etic, and mesenchymal cells. Binding of TGF- to type I and type cancer.

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 5


Iovanna et al. Pancreatic cancer

INTRACELLULAR SIGNALING PATHWAYS INVOLVED IN THE Akt SIGNALING


PANCREATIC CANCER EGF binding and subsequent EGFR, ras, or Src activation lead to
THE EGFR SIGNALING the activation of the PI3K pathway. Activated PI3K phosphorylates
EGFR consists of an extracellular ligand-binding domain, a phosphatidylinositides (PIP3), which eventually phosphorylate
hydrophobic transmembrane region, and an intracellular tyro- and activate Akt which in turn has multiple downstream tar-
sine kinase domain. EGFR is a member of the ErbB family of gets, including the mammalian target of rapamycin (mTOR) and
receptor tyrosine kinases, which include ErbB-1 (EGFR), ErbB- the transcription factor NFB. Phosphorylated Akt (p-Akt) pro-
2 (HER-2), ErbB-3, and ErbB-4. The principal ligands of EGFR motes cell survival by inhibiting apoptosis and activating NFB.
are EGF and TGF-. Binding of a ligand to EGFR induces recep- p-Akt is known to inhibit apoptosis through its ability to phos-
tor dimerization, which results in intracellular transphosphory- phorylate and inactivate several targets including Bad, Forkhead
lation of tyrosine residues. Phosphorylation of EGFR activates transcription factors, and caspase-9, all of which are involved in
molecules in different cell signaling pathways including PI3K, the apoptotic pathway. Akt also regulates the NFB pathway via
Src, MAPK, and STAT, inducing cell cycle progression, cell divi- phosphorylation and activation of molecules in the NFB path-
sion, survival, motility, invasion, and metastasis. The genomic way, suggesting that there is a cross talk between these two signaling
alterations of EGFR that occur in cancers include overexpres- pathways. AKT2 is amplied and the PI3KAkt pathway is acti-
sion, mutation, deletion, and rearrangement. These alterations vated in 20 and 60% of pancreatic cancers, respectively (Ruggeri
of EGFR induce the activity of tyrosine kinase receptors and et al., 1998; Schlieman et al., 2003). Deregulation of this pathway
may promote the development and progression of pancreatic through aberrant expression of PTEN (phosphatase and tensin
cancer. Experimental studies have shown that EGFR activation homolog, a natural antagonist of PI3K) is frequently observed in
plays important roles in proliferation, apoptosis inhibition, angio- pancreatic cancer (Asano et al., 2004). Furthermore, an architec-
genesis, metastasis, and resistance to chemotherapy or radia- tural transcription factor, HMGA1, is overexpressed in pancreatic
tion therapy. Overexpression of EGF and EGFR is a common cancer (Abe et al., 2000). This transcription factor activates PI3K
feature of human pancreatic cancer (Talar-Wojnarowska and Akt signaling and seems to mediate resistance to gemcitabine (Liau
Malecka-Panas, 2006). HER-2/neu amplication and p185 over- and Whang, 2008), which, therefore, provides another target for
expression have been observed in more than 50% of pancreatic inhibition therapy (Trapasso et al., 2004; Liau et al., 2006). Fur-
cancers. Moreover, EGFR overexpression has been found sig- thermore, the inhibition of Akt decreases the function of NFB,
nicantly more often in advanced clinical stages of pancreatic and has been shown to sensitize MiaPaCa2 pancreatic cancer cells
cancer and thus is associated with shorter survival in pancreatic to chemotherapy (Fahy et al., 2004), suggesting that both Akt and
cancer patients (Talar-Wojnarowska and Malecka-Panas, 2006), NFB are potential targets for the treatment of pancreatic cancers.
suggesting that deregulation of the EGFR pathway participates Temsirolimus is an mTOR inhibitor approved for the treat-
in the development and progression of pancreatic cancer. The ment of renal-cell carcinoma, but use of this agent in pancreatic
EGFR signaling and its downstream signaling is therefore an cancer has been limited. Other agents, including everolimus and
important signaling pathway to be targeted for pancreatic cancer sirolimus, are currently in phase II clinical trials (Wolpin et al.,
therapy. 2009). A combination of an mTOR inhibitor with other standard
Two classes of EGFR inhibitors, monoclonal antibodies, and or targeted therapies might be needed, as mTOR expression does
small molecule tyrosine kinase inhibitors have been considered in not correlate with survival of patients.
the treatment of pancreatic cancer but results of comparative clin-
ical trials have been relatively disappointing (see above). Because THE NFB SIGNALING
of the importance of EGFR in pancreatic cancer a novel negative The NFB signaling pathway plays important roles in the con-
regulator of EGFR, termed EGFR-related protein (ERRP), whose trol of cell growth, differentiation, apoptosis, inammation, stress
expression was found to attenuate EGFR activation was devel- response, and many other physiological processes in cellular sig-
oped. ERRP signicantly inhibits cell proliferation and induces naling (Karin, 2006). In human cells without specic signal, NFB
apoptosis in BxPC3, HPAC, and Panc1 pancreatic cancer cells is sequestered in the cytoplasm through tight association with its
(Zhang et al., 2005, 2006). ERRP also inhibits ligand-induced acti- inhibitors: IB which acts as an NFB inhibitor and p100 proteins
vation of EGFR, HER-2, and HER-3. Most importantly, ERRP which serves as both an inhibitor and precursor of NFB DNA-
was found to inhibit pancreatic tumor growth in a SCID mouse binding subunits. NFB can be activated through phosphorylation
xenograft model. The anti-tumor activity of ERRP correlated of IB by IKK and/or phosphorylation of p100 by IKK, leading
well with down-regulation of NFB, MAPK, Akt, and Notch- to degradation of IB and/or the processing of p100 into a smaller
1 (Zhang et al., 2005, 2006; Wang et al., 2006b). ERRP also form (p52). This process allows two forms of NFB (p50p65 and
down-regulates genes downstream from NFB such as vascular p52RelB) to become free, resulting in the translocation of active
endothelial growth factor (VEGF) and MMP-9, and inhibits can- NFB into the nucleus for binding to NFB-specic DNA-binding
cer cell invasion, suggesting that ERRP could be a very potent sites and, in turn, regulating gene transcription. By binding to the
agent for the treatment of pancreatic cancer by inhibiting cell sur- promoters of target genes, NFB controls the expression of many
vival signaling, tumor growth, invasion, and angiogenesis. These genes [i.e., survivin, MMP-9, urokinase-type plasminogen activa-
results suggest that further development of ERRP for clinical use tor (Upa), and VEGF] that are involved in cell survival, apoptosis,
is mandatory. invasion, metastasis, and angiogenesis.

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 6
Iovanna et al. Pancreatic cancer

NFB is constitutively activated in most human pancreatic can- involves the use of gastrimmune, an immunogen that stimu-
cer tissues and cell lines but not in normal pancreatic tissues and lates the formation of antibodies against gastrin 17 and its pre-
cells, suggesting that the activation of NFB is involved in the car- cursors. This agent was, however, not successful in a phase III
cinogenesis of pancreas. Inhibition of NFB by a super-inhibitor trial.
of NFB results in impaired proliferation and induction of apop-
tosis (Liptay et al., 2003), suggesting an important role of NFB
THE HEDGEHOG SIGNALING
in pancreatic tumorigenesis. Moreover, the inhibition of consti-
Hedgehog (Hh) signaling is an essential pathway for embryonic
tutive NFB activity completely suppresses metastasis in liver of
pancreatic development. Hh signaling plays important roles in
the pancreatic cancer cell line ASPC1 (Fujioka et al., 2003). An
adequate tissue morphogenesis and organ formation during gas-
experimental study also demonstrates that uPA, one of the critical
trointestinal tract development. Hedgehog ligands including sonic
proteases involved in tumor invasion and metastasis, is overex-
hedgehog (Shh) are expressed throughout the endodermal epithe-
pressed in pancreatic cancer cells, and its overexpression is induced
lium at early embryonic stages but excluded from the region that
by constitutive NFB activity (Wang et al., 1999). These results
forms the pancreas. Deregulation of the Hh pathway has been
suggest that constitutively activated NFB is tightly related to the
implicated in a variety of cancers including pancreatic cancer.
invasion and metastasis frequently observed in pancreatic cancers.
Overexpression of Shh was reported to contribute to pancre-
Moreover, the deregulation of NFB could also be due to Notch sig-
atic tumorigenesis. Thayer et al. (2003) found that no Shh was
naling as discussed earlier and as such the crosstalk between Notch
detected in the islets, acini, or ductal epithelium of normal pan-
and NFB appears to be an important signaling event that regulates
creas while Shh was aberrantly expressed in 70% of specimens
the processes of tumor invasion and angiogenesis in pancreatic
from patients with pancreatic adenocarcinoma, suggesting that
cancer.
Shh is a mediator of pancreatic cancer tumorigenesis. Importantly,
De novo resistance to chemotherapeutic agents is a major
the down-regulation of Shh by cyclopamine, a specic inhibitor
cause of treatment failure in pancreatic cancer, which could
of Shh, can reduce the growth and viability of pancreatic cancer
be due to the constitutive activation of NFB among others.
cells, suggesting that targeting Shh signaling may be an effective
Moreover, chemotherapeutic agents can activate NFB in pan-
novel approach for the treatment of pancreatic cancer (Berman
creatic and other cancer cells, leading to cancer cell resistance to
et al., 2003). Cyclopamine can enhance sensitivity to radiother-
chemotherapy (acquired resistance). Therefore, the strategies by
apy and chemotherapy and suppress metastatic spread (Shafaee
which NFB could be inactivated represent a novel approach for
et al., 2006; Feldmann et al., 2007) as well as improving anti-
the treatment of pancreatic cancer. Some studies have demon-
tumor activity when combined with an EGFR inhibitor (Hu et al.,
strated that the anti-tumor effects of chemotherapeutic agents
2007). A downstream target of the Shh pathway, the transcrip-
can be enhanced by combination treatment involving inhibi-
tion factor GLI 1, can also be inhibited by miRNA (Tsuda et al.,
tion of NFB. It was shown that NFB activity was signicantly
2006).
increased by cisplatin, docetaxel, doxorubicin, and gemcitabine
A recent report shows that NFB contributes to Hh signaling
treatment. When the NFB inducing activity of these agents was
pathway activation through Shh induction in pancreatic cancer
completely abrogated by pre-treatment of the pancreatic cancer
(Nakashima et al., 2006), demonstrating a crosstalk between NFB
cells with NFB inhibitors, growth inhibition, and apoptosis were
and Hh signaling pathways in pancreatic cancer.
clearly enhanced (Muerkoster et al., 2003; Zhang et al., 2003; Li
et al., 2004b, 2005; Banerjee et al., 2005; Mohammad et al., 2006).
Recently, it was also shown that anti-tumor and anti-metastatic OTHER MECHANISMS INVOLVED IN THE PATHOGENESIS OF
activities of docetaxel could be enhanced by NFB inhibitors PANCREATIC CANCER
through regulation of osteoprotegerin/receptor activator of NFB INFLAMMATION
(RANK)/RANK ligand/MMP-9 signaling in vitro and in vivo (Li There is a growing feeling that inammation contributes to
et al., 2006). pancreatic cancer development. The initiation, promotion, and
progression of tumors may be inuenced by numerous compo-
THE GASTRIN AND CHOLECYSTOKININ RECEPTORS nents including cytokines such as TNF-, IL-6, IL-8 and inter-
The peptide hormone gastrin is secreted by G cells in the gas- feron , the COX-2, and the peroxisome proliferator-activated
tric antrum and duodenum and it can act as a growth factor receptor-, that function in the inammatory response. Other
for gastric, colonic, and pancreatic cancers. CCK-BR (the gas- mechanisms involved in the inammatory response may also
trin and cholecystokinin receptor), gastrin precursors, and the contribute to neoplasia, such as free radicals which may partic-
fully amidated gastrin are expressed in 95, 5591, and 23% of ularly contribute to cancer initiation. Free radicals can induce
pancreatic cancers, respectively (Caplin et al., 2000). A selective genetic alterations and post-translational modications of key
CCK-BR antagonist, gastrazole, was tested in two small, random- cancer-related proteins. Reactive oxygen intermediates and reac-
ized, controlled trials in patients with advanced pancreatic cancer tive nitrogen intermediates are capable of causing oxidative dam-
(Chau et al., 2006). Gastrazole was superior to placebo, but not age and nitration of DNA bases, thus increasing the likelihood
to 5-uorouracil. Another inhibitor, the orally active Z-360, has that DNA mutations occur (Hussain et al., 2003). Interestingly,
demonstrated promising laboratory results and, in combination recognized risk factors for pancreatic cancer such as cigaret
with gemcitabine, is well-tolerated by patients (Kawasaki et al., smoking, chronic hereditary pancreatitis, obesity, and type II
2008). An alternative approach to a blockade of this pathway diabetes are linked by the fact that inammation signicantly

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 7


Iovanna et al. Pancreatic cancer

drives these pathologies. By itself, chronic pancreatitis is associ- gemcitabine and capecitabine in locally advanced and metastatic
ated with increased risk of pancreatic cancer while obesity and pancreatic cancers (Figure 2).
cigaret smoking require additional co-factors to induce pancre-
atic carcinogenesis. The role of inammation in the promotion ANGIOGENESIS
of pancreatic cancer from PanIN lesions has been experimen- Angiogenesis is essential for solid tumor growth, and is princi-
tally conrmed by Guerra et al. (2007, 2011) using an elegant pally mediated by the VEGF family of proteins and receptors.
mutated K-ras-dependent mouse model in which the animals Stimuli that upregulate VEGF expression include hypoxia, other
develop PanIN lesions spontaneously. In these works, repeated growth factors, and oncogenic proteins such as TGF-, EGF, and
episodes of caerulein-induced acute pancreatitis are able to pro- Ras. VEGF is overexpressed in >90% of pancreatic cancers (Seo
mote cell transformation, apparently by blocking the mutated et al., 2000) and is, therefore, an appealing target for therapy.
K-ras-induced senescence. Activation of STAT3 in pancreas cancer Bevacizumab is a humanized antibody against VEGF approved
is related to tumor growth directly through mechanisms affect- for use in patients with colorectal cancer. However, a phase III trial
ing the tumor and indirectly by modulating tumor-associated in advanced pancreatic cancer failed to show any survival ben-
stroma and the immune system. Activation depends on the phos- et for bevacizumab in combination with gemcitabine (Kindler
phorylation of a conserved tyrosine residue (Y705) by upstream et al., 2010). The AVITA (BO17706) phase III study of patients
kinases, such as Janus kinase 2 (Jak2). Jak2 activation requires with metastatic pancreatic cancer reported that the addition of
activation of the ubiquitously expressed gp130 receptor by spe- bevacizumab to gemcitabine and erlotinib did not signicantly
cic ligands (IL-6, LIF, IL-11, oncostatin M, CNTF, and IL-27). prolong overall survival, although a signicant improvement in
Importantly, it was recently reported that STAT3 activity plays a progression-free survival was seen (Van Cutsem et al., 2009). A
critical role in K-ras-induced pancreatic tumorigenesis (Corcoran number of other trials are being conducted to examine beva-
et al., 2011) supporting the link between STAT3 activation and cell cizumab in combination with other agents or treatment modalities
transformation (Figure 2). for pancreatic cancer; however, this agent seems unlikely to con-
fer sufcient benet to justify its licensing for this condition. The
THE TELOMERASES failure of bevacizumab in therapeutic trials for pancreatic cancer
The telomeres located at the end of chromosomes normally shrink highlighted the need for angiogenic inhibitors that could target
with each cell division and thereby impose a nite lifespan to other non-VEGF pathways and have better access to the tumor
the cell. Most malignant cells have detectable activity of telom- environment than an antibody. Sorafenib is a multitargeted kinase
erase, a reverse transcriptase that contains an RNA template and inhibitor that inhibits the VEGF receptor (VEGFR), PDGFR, SCFR
elongates telomeres. Telomerase is overexpressed in 95% of pan- (formerly c-KIT), Raf1, and FLT3, which are all implicated in
creatic cancers (Hiyama et al., 1997) which provides a rationale tumor growth and angiogenesis. Sorafenib was approved in 2005
for the development of antitelomerase agents. GV1001 is a telom- for the treatment of advanced renal-cell carcinoma. However, a
erase peptide vaccine that has shown some promising results in phase II study concluded that, although well-tolerated, it was
phase I/II studies (Bernhardt et al., 2006). This vaccine is being inactive in patients with advanced pancreatic cancer. A random-
tested in the large (>1000 patients), phase III, TeloVac trial with ized phase III study comparing gemcitabine plus sorafenib versus
gemcitabine plus placebo was recently reported and revealed no
advantage for the experimental arm (Gonalves et al., 2011). Axi-
tinib is an orally active inhibitor of both VEGFR and related tyro-
sine kinase receptors at high concentrations. A median survival of
6.9 months was reported for axitinib combined with gemcitabine
compared with 5.6 months for gemcitabine alone in a phase II
trial in patients with advanced pancreatic cancer, but this nding
was not statistically signicant (Spano et al., 2008). Phase III trials
of axitinib combined with gemcitabine are currently in progress.
Aibercept, a recombinant fusion protein that functions as a solu-
ble decoy receptor and thereby inhibits VEGF, is another novel
agent being tested in a phase III trial of patients treated with
gemcitabine for metastatic pancreatic cancer (Figure 2).

SIGNALING BY INTEGRIN RECEPTORS


Integrin receptors on the cell surface interact with the extracellular
matrix and mediate various signaling pathways. These receptors
are involved in many neoplastic processes, including tumor sur-
vival, invasion, and metastasis. The V3 and V5 integrins
FIGURE 2 | Mechanisms involved in pancreas cancer. At the upper part, induce angiogenesis, principally via basic broblast growth fac-
in red, are shown the mechanisms that promote pancreatic cancer, and at
tor and VEGF, respectively. Cilengitide inhibits these integrins,
the lower part, in green, are showed the mechanisms against pancreatic
cancer development. but in a phase II trial in patients with advanced pancreatic can-
cer it did not show signicant benet compared to gemcitabine

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 8
Iovanna et al. Pancreatic cancer

alone (Friess et al., 2006). Other anti-integrin agents, including (Roldo et al., 2006; Szafranska et al., 2007). Monitoring differ-
an antibody against 51 (volociximab; Ricart et al., 2008) and an ential miRNA expression might therefore be useful in the differ-
inhibitor of 2 (E7820) are in early phase clinical trials (Funahashi ential diagnosis of pancreatic cancer. Expression of the miR-376
et al., 2002; Figure 2). precursor was highest in the human pancreatic cancer cell line
Panc1, compared to other cell lines studied (Jiang et al., 2005;
MATRIX METALLOPROTEINASES Volinia et al., 2006). Large-scale miRNA proling in 540 sam-
Matrix metalloproteinases (MMPs) are a family of zinc-dependent ples of solid tumors (breast, colon, lung, pancreas, prostate, and
proteolytic enzymes that degrade the extracellular matrix and are stomach) showed that the spectrum of miRNA expression varied
essential for tumor spread and neovascularization. We should not, in different solid tumors and was different from that of normal
therefore, be surprised that imbalance between MMPs and their cells (43 of 137 miRNAs, 31%). miR-21, miR-191, and miR-17-5p
natural inhibitors is a frequent event in pancreatic cancer. Despite were signicantly overexpressed in all six tumor types, whereas
promising laboratory results, MMP inhibitors have failed to live up miR-218-2 was consistently down-regulated in colon, stomach,
to their initial therapeutic expectation in three phase III clinical tri- prostate, and pancreatic cancers, but not in lung and breast carci-
als (Bramhall et al., 2001, 2002; Moore et al., 2003), although critics nomas. This observation indicates that colon, pancreas, prostate,
argued that the trials included a large number of patients with and stomach have similar miRNA signatures, different from those
metastatic disease, which contradicts the rationale of exploiting of breast and lung cancer. Similarly, up-regulation of miR-142-3p,
the cytostatic effect of MMP inhibitors (Figure 2). miR-142-5p, miR-155, and miR-146a expressions was observed
in human pancreatic neuroendocrine tumors (PNETs) as com-
CANCER STEM CELLS pared with normal human islets (Olson et al., 2009). Lee et al.
Cancer stem cells possess important properties associated with (2007) reported the aberrant expression of 100 miRNA precur-
their normal counterparts, namely the ability for self-renewal and sors in pancreatic cancer or desmoplasia, including the miRNAs
differentiation. Pancreatic cancer stem cells are identied by their previously reported in other human cancers (miR-155, miR-21,
surface markers, such as CD133, CD44, CD24, and otillin 2 miR-221, and miR-222) as well as the rst reported miR-376a and
epithelial specic-antigen. Evidence suggests that such cells form miR-301 for the differential expression of cancer. A signicant up-
a small subset in the heterogenous tumor population, and con- regulation of miR-196a, miR-190, miR-186, miR-221, miR-222,
tribute to neoplastic progression, metastasis, and resistance to miR-200b, miR-15b, and miR-95 in most pancreatic cancer tissues
chemotherapy and radiotherapy (Hermann et al., 2007; Lee et al., and cell lines was reported. miR-155 and miR-21 were signi-
2008). For this reason, cancer stem cells are thought to be respon- cantly up-regulated in 15 IPMNs versus matched controls (Habbe
sible for relapse of disease after clinical remission. Dysregulation et al., 2009). miR-155 was up-regulated in 53 of 64 IPMNs (83%)
of various signaling cascades, including the PTEN, Shh, Notch, compared to 4 of 54 normal ducts only (7%), and miR-21 was
and Wnt pathways, are frequently observed in cancer stem cells, up-regulated in 52 of 64 IPMNs (81%) compared to 1 of 54 nor-
which provides further rationale for use of these pathways as a mal ducts (2%). The expression of miR-216 has also shown to
target for therapeutic purposes. Further studies are still needed to be specic to the pancreas (Sood et al., 2006). Let-7 miRNA was
understand the genetic and biological properties of cancer stem found expressed in pancreatic cancer cells but did not inhibit the
cells for the development of effective treatment modalities. epithelialmesenchymal transition (Watanabe et al., 2009). There-
fore, miRNAs play vital roles not only in different kinds and stages
miRNA EXPRESSION PROFILING IN PANCREATIC DUCTAL of pancreatic tumors but also in many diseases, including can-
ADENOCARCINOMA cers, cardiovascular diseases, and immune disorders. Epigenetic
Analysis of miRNA expression in PDAC revealed a specic miRNA modications, DNA copy number changes, and genetic mutations
signature that can be monitored by proling miRNAs at different may regulate the expression of miRNAs. The conclusion of these
stages of cancer (Table 1). Over the past years, a number of dif- data is that PDAC miRNAomes associated with normal and tumor
ferent approaches, including DNA microchips and RT-PCR have tissues are different. Differences are tumor-specic and, in some
been described to quantify miRNAs. By these techniques, several cases, indicators of prognosis. These ndings suggest that miRNA
studies demonstrated the tissue-specicity of miRNA expression expression patterns constitute a signature of the disease which
and the deregulation of miRNA expression in pancreatic cancer could offer new clues about pancreatic cancer occurrence and also
provide new molecular markers that would improve diagnosis and
orient the treatment. A very promising diagnostic strategy could
Table 1 | Genes implicated in the familiar forms of pancreas cancer. arise from miRNAs if they are found in serum and can be detected
by RT-PCR. In a remarkable study Lu et al. (2005) showed that
Hereditary breast and BRCA1 and BRCA2
expression data for 217 miRNAs only performed better at identi-
ovarian cancer syndrome
fying cancer types than analysis of 16000 mRNAs. They concluded
PeutzJeghers syndrome LKB1 that miRNAs might help detecting cancer better than other strate-
Hereditary chronic pancreatitis PRSS1 and SPINK1 gies presently available because miRNAs are only several hundred,
Hereditary non-polyposis colorectal hMSH2, hMLH1, hPMS1, compared to tens of thousands for mRNAs and proteins. One sin-
cancer syndrome hPMS2, and hMSH6 gle miRNA can modulate the expression of many genes rendering
Familial atypical multiple mole melanoma CDKN2A/p16 miRNAs powerful molecules regulating several related pathways,
Familial adenomatous polyposis APC each miRNA playing multiple but coherent roles in the cell.

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 9


Iovanna et al. Pancreatic cancer

EPIGENETICS OF PANCREATIC CANCER has been associated with transcriptional repression of tumor sup-
The revolution of somatic genetics in the eld of cancer brought pressor genes that cause growth inhibition and apoptosis (Cress
about by the model developed by Fearon and Vogelstein (1990) in and Seto, 2000). In a study performed by Ouassi et al. (2008)
colon, which later led to an adaptation to the pancreas by Hruban 80% of pancreatic adenocarcinoma samples examined showed
et al. (2000), opened a fruitful era for pancreatic cancer research. a signicant increase in HDAC7 RNA and protein levels (Ouassi
The basic premise of somatic genetics in cancer is that if a gene, et al., 2008). Thus, it is clear that HDACs play an important role
which is suspected to play a role related to cancer, is over-amplied in the maintenance of the proper balance of chromatin marks on
or mutated with gain-of-function, it behaves as an oncogene, but if a given promoter. If this balance is altered, such as with HDAC
it is deleted or mutated with loss-of-function, it behaves as a tumor overexpression in pancreatic cancer, the expected global effect on
suppressor. However, in light of the modern biology we can assume promoters is daunting.
that regulation of genes activity is more complex and it is there-
fore necessary to explain how changes in chromatin dynamics can HISTONE H3METHYL-K27 AND POLYCOMB
silence tumor suppressor genes via mechanisms that are totally Polycomb proteins silence gene expression by specically methy-
independent from either deletions or mutations of these genes. lating histone H3, on K27 (Cao et al., 2002; Ringrose, 2007). At the
Some epigenetic mechanisms can silence gene function such as: core of this pathway, the polycomb group of proteins (PcG) act
via the stepwise recruitment of PRC2, containing the histone H3
DNA METHYLATION K27 methylase activity, to chromatin. Subsequently, the trimethyl-
DNA methylation was the rst type of epigenetic change to be K27H3 mark deposited by PRC2 recruits the PRC1 complex,
studied as a mechanism for the inactivation of tumor suppres- thereby completing the formation of the gene silencing complex.
sors (Esteller, 2008). DNA methylation occurs on dinucleotide The enzymatic activity of the PCR2 complex involves the K27H3
CpGs. The process of DNA methylation entails the addition of a histone methylase, EZH2, but requires formation of a complex
methyl group to carbon number 5 of the cytosine pyrimidine ring, with Suz12 and EED to function. The PCR1 complex contains
which ultimately silences gene expression. In pancreatic cancer, the oncogene BMI1, as well as HPC1-3, HPH1-3, SCMH1, and
DNA methylation has been known for a long time as a mech- the methyl-K27H3-binding proteins, Cbx 2, 4, 6, 7, and 8. The
anism for inactivating tumor suppressor genes, a well known role of polycomb proteins in pancreatic cancer is an emerging
example being the inactivation of the p16 promoter via methy- area of research. Interesting studies have demonstrated that loss
lation (Singh and Maitra, 2007). In addition, loss of methylation of trimethylation at lysine 27 of histone H3, which is achieved by
of a normally silenced promoter in pancreatic cells, such as in EZH2, is a predictor of poor outcome in pancreatic cancers (Wei
the gene encoding the hematopoietic-specic guanine nucleotide et al., 2008). In fact, together with tumor size and lymph node sta-
exchange factor, VAV1, can alter its expression (Fernandez-Zapico tus, the level of trimethyl-K27H3 was found to have a strong and
et al., 2005). Although several genes were discovered to be methy- independent prognostic inuence in pancreatic cancer. In another
lated in advanced pancreatic cancer, current evidence supports recent study, nuclear accumulation of EZH2 was identied as a
the idea that aberrant methylation occurs very early during the hallmark of poorly differentiated pancreatic adenocarcinoma and
histopathological progression of this neoplasia. Interestingly, in nuclear overexpression of EZH2 contributes to pancreatic can-
a study involving large-scale methylation analysis, Sato et al. cer cell proliferation, suggesting EZH2 as a potential therapeutic
(2003) analyzed DNA samples from 65 PanIN lesions for methy- target for the treatment of pancreatic cancer (Ougolkov et al.,
lation status of eight genes, identied beforehand by a large-scale 2008). Mechanistically, one of the outcomes of aberrant poly-
microarray approach as aberrantly hypermethylated in invasive comb regulation is the silencing of the p16 gene, which could
pancreatic cancer. Methylation of these genes was identied in 68% occur prior to DNA methylation, directly via altered recruitment
of PanINs. More importantly, aberrant methylation was present in of members of this family to the p16 promoter sequence (Kotake
approximately 70% of PanINs-1A, the earliest lesions. The preva- et al., 2007). Upon recent studies in human cells, EZH2, and DNA
lence of methylation increased from PanIN-1 to PanIN-2 for the methyltransferases (DNMTs) were found to physically and func-
NPTX2 gene, and from PanIN-2 to PanIN-3 for SARP2, Reprimo, tionally interact, evidenced by the PRC2 subunits, EZH2, and
and LHX1. EED, co-immunoprecipitating with all three human DNMTs and
the co-dependency of certain target gene silencing requiring both
HISTONE ACETYLATION AND DEACETYLATION EZH2 and DNMTs (Vire et al., 2006). Therefore, the presence of
Another important mechanism underlying the epigenetic reg- polycomb proteins on the p16 promoter can recruit DNA methy-
ulation of gene expression is the acetylation and deacetylation lases which then further inactivate the expression of p16 via DNA
of lysine residues within histone tails. Regarding acetylation, the methylation.
process occurs via HATs, such as CBP, P300, and PCAF, to result in
activation of gene expression, whereas deacetylation is mediated HISTONE H3METHYL-K9 AND HETEROCHROMATIN PROTEIN 1
by two different families of HDACs, resulting in gene silenc- HP1 binds to methylated K9 of histone H3, causing transcrip-
ing. Together, these enzymes provide a ne-tuned mechanism tional repression (Lomberk et al., 2006). This occurs through
which, upon alteration, can promote the activation of oncogenic the N-terminal chromodomain of HP1, while the highly related
pathways and the silencing of tumor suppressors. Studies have C-terminal chromo shadow domain allows for dimerization of
demonstrated that HDAC activity is increased in various tumors these HP1 molecules and serves as a docking site for various fac-
compared with normal tissue, and this increase in HDAC activity tors involved in a wide array of functions, from transcription to

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 10
Iovanna et al. Pancreatic cancer

maintaining nuclear architecture. To mediate gene silencing via GENETICS OF THE FAMILIAL PANCREATIC
the formation of heterochromatin, HP1 isoforms must interact ADENOCARCINOMA
with two different H3K9 histone methylases namely G9a and In addition to sporadic adenocarcinoma, an increased risk of pan-
Suv39h1 (Lomberk et al., 2006). These methylases work in con- creatic cancer has been demonstrated among persons with a family
cert with HP1 in a circular manner to form silenced chromatin. history of pancreatic cancer. Coughlin et al. (2000) reported an
When either of the methylases adds methyl groups to K9, this, in increased risk of developing pancreatic cancer for individuals who
turn, forms an HP1 docking site on chromatin. Since HP1 also reported a positive family history of pancreatic cancer at baseline,
recruits the methylases, this cycle repeats, and the HP1-methylase with a relative risk of 1.5 after adjustment for age. Furthermore,
pair can spread the formation of silenced chromatin to adjacent a population-based cohort study demonstrated that the risk of
nucleosomes, causing long-term silencing of entire genes. pancreatic cancer increased 1.72-fold for individuals with a par-
ent who developed a pancreatic cancer. The risk was not elevated
EPIGENETICS AS TARGET FOR CHEMOPREVENTION AND when a more distant relative had been diagnosed with pancre-
CHEMOTHERAPIES atic cancer. Thus, several studies strongly support the hypothesis
The study of epigenetics also impacts the eld of therapeu- that familial aggregation and genetic susceptibility play an impor-
tics. In stark contrast with genetic alterations, epigenetic changes tant role in the development of pancreatic cancer. Discovering the
have been proven to be reversible. For instance, methylation genetic basis of inherited pancreatic cancer is an active area of
can be reversed through the use of 5-Aza-2 -Deoxycytidine or research. In 2001, a multicenter linkage consortium, PACGENE,
DNA methyltransferase inhibitors. Two types of DNMT inhibitors was established to conduct linkage studies aiming at localizing
exist, namely, nucleoside and non-nucleoside (small molecule) and identifying pancreatic cancer susceptibility genes (Petersen
inhibitors (Mund et al., 2006; Grifths and Gore, 2008). These et al., 2006). Other groups have used linkage studies to suggest
types of pharmacologic DNMT inhibitors are currently being that mutations in the paladin gene (PALD) on chromosome 4q32
tested in phase IIII clinical trials. Currently, among the most predisposes to pancreatic cancer (Pogue-Geile et al., 2006); how-
promising epigenetic treatments for cancer are the potential ther- ever, this nding has not been validated in subsequent studies (Earl
apeutic agents that target histone deacetylation HDAC inhibitors et al., 2006). The complex nature of pedigree data makes it dif-
(HDACIs; Berger, 2002; Glaser et al., 2003; Bruserud et al., 2007; cult to accurately assess risk based upon the simple counting of
Hildmann et al., 2007; Pan et al., 2007). Intuitively, the proposed the number of affected family members, as it does not account
mechanisms of action for HDACs attempt to harness their ability for family size, current age, or age of onset of pancreatic can-
to reactivate transcription of multiple genes found to be silenced cer, and the exact relationship between affected family members.
in human tumors. However, their pleiotropic anti-tumor effects, Computer-based, risk assessment tools have been developed to
specic for cancer cells, along with their potential activity in other integrate this complex risk factor and pedigree data into risk assess-
diseases, elicits the possibility that they also act in other biolog- ment. These models can provide more precise risk assessment than
ical processes, currently poorly understood. In addition to being guidelines or models that rely on counts of affected family mem-
well-tolerated, several HDACIs show promising anti-tumor activ- bers, such as the Bethesda Guidelines for hereditary non-polyposis
ity with more than 50 naturally occurring or synthetic HDACIs colorectal cancer (HNPCC) or myriad tables for hereditary breast
currently developed. Hydroxamic acid compounds, trichostatin A and ovarian cancer. In April 2007, the rst risk prediction tool
(TSA), and suberoylanilide hydroxamic acid (SAHA), are the best for pancreatic cancer, PancPRO, was released (Wang et al., 2007).
known among these agents. By the same token, however, other Although the genetic basis for most instances of aggregation of
equally promising, although less known, drugs can be classied pancreatic cancer in families is unknown, the genes responsible for
into wider groups such as short-chain fatty acids (e.g., valproic a small portion of familial pancreatic cancer are known (Table 2).
acid), epoxides (e.g., trapoxin), cyclic peptides (e.g., apicidin), ben- Germline mutations in the BRCA2, CDKN2A/p16, STK11/LKB1,
zamides (e.g., CI-994, Nacetyldinaline), and hybrid compounds and PRSS1 genes have all been shown to increase the risk of pancre-
(e.g., SK-7068). Recent studies have shown that certain HDACIs atic cancer (Lowenfels et al., 1993; Goldstein et al., 1995; Whitcomb
can induce death of cultured pancreatic cells (Ouassi et al., 2008). et al., 1996; Giardiello et al., 2000). Additionally, some studies have
Additionally, conventional chemotherapeutic drugs, such as gem- described pancreatic cancers developing among individuals with
citabine, in combination of some of these agents, can achieve a HNPCC; however, the association between HNPCC syndromes
synergistic inhibition of pancreatic adenocarcinoma cell growth and pancreatic cancer is not as well-dened as it is for some of the
(Donadelli et al., 2007). As a result of these promising studies, new other syndromes (Abraham et al., 2001; Yamamoto et al., 2001).
agents belonging to this family of drugs, such as FR235222, could While most patients with sporadic and familial pancreatic cancer
potentially be used as therapeutic tools in this cancer (Singh et al., have classic inltrating ductal (tubular) adenocarcinoma, some
2008). HDACIs are in advanced phases of clinical trials and may inherited syndromes are associated with a specic histologic type.
soon become part of the therapeutic regimen for oncologists treat- Although rare, these cases provide a unique opportunity to corre-
ing this disease. It is interesting to note that HDACIs are already late genetics with histology. For example, many pancreatic cancers
potential therapeutic agents against pancreatic cancer, in spite of that develop in patients with HNPCC syndrome have a medullary
the fact that the characterization of histone deacetylases in pancre- phenotype (Goggins et al., 1998a; Wilentz et al., 2000; Banville
atic cancer is far from being completed and their role is not fully et al., 2006) and individuals with the PeutzJeghers syndrome
understood. appear to be predisposed to IPMNs (Su et al., 1999; Furukawa,

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 11


Iovanna et al. Pancreatic cancer

Table 2 | microRNAs frequently misregulated in pancreatic cancers. pancreatic cancer cohort consortium, or PCCC, an international
consortium comprising Centers from Europe, China, and North
Up Down America. In these studies, 12 cohorts representing more than 3800
cases of cancer and 3900 controls allowed detection of four new
miR-155 miR-375
genes associated with pancreatic cancer. The rst locus, harbor-
miR-100 miR-345
ing several SNPs strongly associated with cancer occurrence, was
miR-376a miR-142-P
localized on chromosome 9q34 and contains the rst intron of
miR-125b-1 miR-139
the ABO gene. That gene encodes the glycosyl-transferase which
miR-181a miR-148a
catalyzes the transfer of sugars to antigen H (Histo-blood group
miR-181c miR-148b
ABO system transferase). By this mechanism, antigen H becomes
miR-146a miR-141
antigen A or B, depending on the nature of the sugar transferred by
miR-196a miR-96
the ABO enzyme. In individuals from group O, the gene is mutated
miR-25 miR-29c
and generates a non-functional protein. Association of the ABO
miR-214 miR-130b
gene with pancreatic cancer has already been suggested 50 years
miR-222 miR-216
ago in studies showing a higher frequency of pancreatic or gastric
miR-29b-2 miR-217
cancer in individuals from groups A, B, or AB than from group O
miR-128b miR-107
(Aird et al., 1953; Marcus, 1969). That gene is also found altered
miR-200 miR-34a
in primary tumors and metastases of pancreatic cancer (Itzkowitz
miR-95 Let-7
et al., 1987). More recently, the PCCC added three other regions
miR-15b miR-218-2
strongly associated with cancer (Petersen et al., 2010). The rst
miR-32
one is an intergenic region of 600 kb in chromosome 13q22.1,
miR-30c
located between genes KLF5 and KLF12. These two members of
miR-21
the Kruppel transcription factor family are involved in regulating
miR-17-92
cell growth and transformation (Dong and Chen, 2009; Naka-
miR-191
mura et al., 2009). The second region is located on 1q32.1. That
miR-221
region harbors gene NR5A2 (Nuclear Receptor subfamily 5 group
miR-190
A member 2), 91 kb upstream from the gene. The receptor encoded
miR-186
by gene NR5A2 is present, in adults, in the exocrine pancreas, liver,
gut, and ovaries. Its function is not completely understood. In pan-
2007). These associations between phenotype and genotype are creas, NR5A2 would contribute to the regulation of the expression
important because tumor phenotype can be used to identify at-risk of several genes (Fayard et al., 2003). The last locus identied by
families. this consortium is localized at 5p15.33. SNP markers that border
this locus are within intron 13 of the CLPTM1L gene (Cleft lip and
GERMLINE DNA MUTATIONS THAT INCREASE palate transmembrane 1-like). CLPTM1L is poorly described. One
SUSCEPTIBILITY TO DEVELOP PANCREATIC of the critical factors in the interpretation of GWAS is the ethnicity
ADENOCARCINOMA of the studied group. PCCC studies mentioned above have been
Most pancreatic cancers are sporadic. In sporadic forms, asso- conducted on Caucasian populations and do not necessarily reect
ciation with polymorphic somatic mutations, spontaneous, or risk factors for other ethnic groups. In fact, a GWAS conducted by
generated by environmental factors is of paramount importance. Biobank Japan (BJJ) has evidenced a different set of markers of
It is well known that sporadic forms of cancers result from the pancreatic cancer (Low et al., 2010). Among loci strongly associ-
accumulation of genetic modications or mutations. Besides par- ated with the disease, three are specic to the Japanese population.
ticular syndromes whose association with pancreatic cancer is well The rst one, in 6p25.3, is a 75-bp linkage disequilibrium block
known, a lot of work remains to identify genetic variations asso- containing the FOXQ1 gene [Forkhead-box (Fox) Q1]. The second
ciated with cancer predisposition. Recent advances in large-scale SNP showing signicant association is located within gene BICD1
genotyping (SNP chips) increased considerably the number of (Bicaudal-D homolog 1). The last locus with a high odd-ratio cor-
markers that can be simultaneously genotyped. As a consequence, responds to several SNPs that fall in the rst intron of the DPP6
haplotype blocks can be determined much more precisely than gene (Dipeptidyl-peptidase 6, a member of the DPP family devoid
with microsatellite markers. These methodologies allowed recent of the catalytic residues required for enzymatic activity but which
studies of associations covering all intragenic regions of the whole acts through proteinprotein interactions).
human genome, these studies being called genome-wide associ- Two studies have extended the results obtained by analyzing
ation studies (GWAS). Several GWAS led to the identication of tumor exome, including a study comparing the exomes of the pri-
susceptibility markers of several types of cancers (review in Mano- mary tumor and corresponding metastases (Campbell et al., 2010;
lio, 2010). More than 700 GWAS are presently registered but only Yachida et al., 2010). In the rst one, Yachida et al. (2010) gathered
four concern pancreatic cancer (Amundadottir et al., 2009; Dier- data on the exomes of seven tumors previously characterized by
gaarde et al., 2010; Low et al., 2010; Petersen et al., 2010). Two Jones et al. (2008), and compared them to data on the exomes of
GWAS for pancreatic cancer (Amundadottir et al., 2009; Petersen the corresponding metastases. The second study was conducted on
et al., 2010), have been conducted within the framework of the 13 tumors and corresponding metastases (Campbell et al., 2010).

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 12
Iovanna et al. Pancreatic cancer

Information produced by these studies allowed to ll in a high in pancreatic cancer cells (Li et al., 2004a). Phase II trials of cur-
resolution framework of the various modications and rearrange- cumin with and without gemcitabine showed that curcumin was
ments that occur in the DNA of metastases, of the various underly- well-tolerated and might have some biological activity in patients
ing mechanisms, on the genes potentially involved in the metastatic with pancreatic cancer (Dhillon et al., 2008).
process and nally on the time between the appearance of the can-
cerous cell and the occurrence of a clone with a metastatic prole. SOY ISOFLAVONES
Genomic instability is apparently the main characteristic of pan- Genistein is the main isoavone found in a relatively high con-
creatic cancer (Campbell et al., 2010). DNA sequencing in tumors centration in soybeans and most soy-protein products. Genis-
and in cells at different stages of progression toward metastases tein inhibits cell growth and induces apoptosis in various can-
revealed that the major chromosomal rearrangement is a dupli- cers including pancreatic cancer. Genistein treatment signi-
cation of fragments and their insertion in opposite directions, a cantly inhibited NFB DNA-binding activity, inhibited Akt activity
phenomenon called fold-back inversions by the authors. The (Banerjee et al., 2005; Li et al., 2005) and signicantly down-
origin of fold-back inversions is not known. A possible hypothe- regulated Notch signaling, leading to the inhibition of NFB and
sis is that early in tumor progression telomerase activity is altered induction of apoptosis in pancreatic cancer cells (Wang et al.,
or inhibited (Bardeesy and DePinho, 2002) generating a progres- 2006c,e).
sive shortening of telomeres and successive cycles of breaking and
abnormal fusion of chromatids by a mechanism called breakage INDOLE-3-CARBINOL AND 3,3 -DIINDOLYLMETHANE
fusionbridge (McClintock, 1941). These rearrangements will Indole-3-carbinol (I3C) is produced from all kinds of cabbages,
determine the gain or loss of DNA and, as consequence, genomic broccoli, cauliower, and Brussels sprouts. I3C is biologically
instability. Interestingly, these results suggest that fold-back inver- active and it is easily converted in vivo to its dimeric product 3,3 -
sions arise early in tumor evolution and would trigger progression diindolylmethane (DIM), which is also biologically active. Both
toward metastasis (Campbell et al., 2010). These results have I3C and DIM inhibit cell proliferation and induce apoptotic cell
shown unequivocally, for the rst time, that the pancreatic tumor death in a variety of cancers including pancreatic cancer (Abdel-
evolves from a rst (parental) clone which generates progres- rahim et al., 2006). I3C induces apoptosis in pancreatic cancer
sively other sub-clones. Each sub-clone will evolve independently cells through the inhibition of STAT3 whose activation has been
by accumulating specic genetic modications and, possibly, a observed in human pancreatic carcinoma specimens and pancre-
tissue-specic metastatic afnity prole. Establishment of a phy- atic cell line but not in normal pancreatic tissues (Lian et al.,
logenetic correlation between genomic alterations of tumors and 2004).
their metastases allowed drawing a kinetic of tumor progression
(Yachida et al., 2010). Unexpectedly, the mean time between aris- GREEN TEA
ing of the founding cell and the occurrence of the rst parent clone Consumption of green tea has been implicated in better human
(non-metastasis) is 11.8 years. Another 6.8 years will be required health including the prevention of cancers. Green tea con-
for one of the sub-clone to acquire the metastatic prole, which tains several catechins including epicatechin (EC), epigallocate-
occurs about 3 years before the patient dies. These ndings, if con- chin (EGC), epicatechin-3-gallate (ECG), and epigallocatechin-
rmed, have major clinical consequences. They show that, if early 3-gallate (EGCG). However, EGCG is among catechins the most
detection of pancreatic cancer is possible, there is a large window potent for oncogenesis inhibition and reduction of oxidative stress
of about 10 years for therapeutic intervention. (Mukhtar and Ahmad, 1999). By targeting multiple signaling path-
ways including MAPK, EGFR, and NFB, EGCG inhibits the
PANCREATIC CANCER PROTECTION BY DIETARY malignant transformation of epidermal cell lines, inhibits cell
CHEMOPREVENTIVE AGENTS growth, and induces apoptosis in a number of cancer cells includ-
More than two-thirds of human cancers could be prevented ing pancreatic cancer cells (Mukhtar and Ahmad, 1999; Qanungo
by modication of lifestyle including dietary modication. The et al., 2005; Khan et al., 2006).
dietary factors associated with increased risk of pancreatic cancer
are meat, red meat in particular, and energy. Protection is mainly RESVERATROL
provided by fruit, vegetables, and vitamins. In recent years, more Relatively high quantities of resveratrol are found in grape juice
dietary compounds have been recognized as cancer chemopre- and red wine. Resveratrol has been shown to have benecial effects
ventive agents because of their anti-carcinogenic activity. There- on the reduction of oxidative stress and the prevention of cancers.
fore, early invasion and metastasis of pancreatic cancer could be Resveratrol was rst noted to be a cancer chemopreventive agent
preventable by these dietary compounds. In addition to dietary having antioxidant and anti-tumorigenic properties (Jang et al.,
factors, there are some chemopreventive agents such as: 1997). Like EGCG, it is a polyphenol which can cause cell cycle
arrest in G1 in various tumor cell lines including pancreatic cancer
CURCUMIN (Ding and Adrian, 2002).
Curcumin is a compound from Curcuma longa (tumeric). Cur-
cumin has recently received considerable attention due to its pro- LYCOPENE AND VITAMINS
nounced anti-inammatory, anti-oxidative, immunomodulating, Tomato products including ketchup, tomato juice, and pizza sauce,
anti-atherogenic, and anti-carcinogenic activities. Curcumin sup- are the richest sources of lycopene in the occidental diet. Dietary
presses the activation of NFB through inhibition of IKK activity intake of lycopene is associated with reduced pancreatic cancer

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 13


Iovanna et al. Pancreatic cancer

risk, suggesting a role in the prevention of pancreatic cancer Gemcitabine treatment was associated with improvement in the
(Nkondjock et al., 2005). median overall survival (5.65 versus 4.41 months, P = 0.0025) and
1-year survival (18 versus 2%). Following this trial, several stud-
CURRENT TREATMENTS ies have evaluated in phase III randomized trials the impact of
Due to delayed diagnosis and aggressiveness of pancreatic cancer, adding a second cytotoxic compound to gemcitabine, notably 5-
only 10% of patients are eligible for curative treatment and 90% of FU and analogs (such as capecitabine, an oral 5-FU pro-drug;
patients undergo palliative therapies. Surgery, chemotherapy, and Berlin et al., 2002; Herrmann et al., 2007; Cunningham et al.,
radiotherapy are the weapons used to ght pancreatic cancer, with 2009) or platinum (cisplatin or oxaliplatin) derivatives (Colucci
disappointing impact on survival though. et al., 2002, 2010; Louvet et al., 2005; Heinemann et al., 2006;
Poplin et al., 2009). No clear survival advantage was demonstrated
SURGERY in these trials, even though some of them have suggested a bene-
To date, surgery remains the most efcient treatment. However, t in patients with good performance status (Heinemann et al.,
only 10% of patients are eligible for resection: no distant dis- 2008). Several classes of targeted agents have also been tested
ease (hepatic or lung metastasis) and no local vascular invasion in combination with gemcitabine and were compared to gemc-
(mainly mesenteric vessels and celiac trunk) must be present to itabine alone. In a phase III trial, the combination of erlotinib,
consider curative resection. Palliative surgery can be performed in an orally administered EGFR tyrosine kinase inhibitor, plus gemc-
selected patients but is not recommended: endoscopic derivation itabine versus gemcitabine alone showed statistically signicant
(duodenal prosthesis and biliary duct stenting) has reduced the improvements in overall survival [6.2 versus 5.9 months; haz-
eld of the historical twin derivation. The last two decades have ard ratio (HR): 0.82; P = 0.038] and 1-year survival (23 versus
shown a dramatic reduction in postoperative morbidity and mor- 17%, P = 0.023), leading to its approval by health authorities
tality of pancreatic surgery. Now, pancreatic surgery performed for rst-line treatment of patients with advanced pancreatic can-
by experienced surgeons in high volume centers results in less cer. However, the small improvement in survival, only 2 weeks,
than 5% mortality. Due to reduced morbidity, pancreatic surgery was associated with increased toxicity and therefore not consid-
can be performed safely in elderly patients. Moreover, vascular ered as clinically meaningful (Moore et al., 2007), explaining why
resection/reconstruction (only recommended for venous involve- it is barely used in the routine setting. Cetuximab, a chimeric
ment; arterial resection remains a contra indication to surgery) monoclonal antibody targeting EGFR, was also evaluated in com-
is becoming safe and increases the proportion of patients elected bination with gemcitabine versus gemcitabine alone, but no dif-
for pancreatic surgery. Despite appropriate surgery and perioper- ference in term of response, progression-free survival, or overall
ative treatments, the 5-year overall survival after resection remains survival was observed (Philip et al., 2010). Two phase III trials
poor and long-term survivors are rare. Yet, some progress has been investigated the potential impact of bevacizumab, a humanized
made. In the 80s, a new type of pancreatic tumor was identied and monoclonal antibody targeting VEGF, in advanced pancreatic can-
named IPMNs (Sohn et al., 2001). IPMN is dened by a benign cys- cer patients (gemcitabine plus bevacizumab versus gemcitabine
tic tumor connected with the pancreatic duct network. However, alone; gemcitabine plus erlotinib plus bevacizumab versus gem-
transformation in pancreatic cancer occurs in 520% of IPMN citabine plus erlotinib plus placebo) but failed to demonstrate
according to sub-IPMN types (side branch or main duct IPMNs). any overall survival advantage (Van Cutsem et al., 2009; Kindler
Thus, patients diagnosed with IPMN (fortuitous diagnosis, pan- et al., 2010). Other tested targeted agents to date, such as tip-
creatitis, etc.) had to be strictly followed up or their pancreas ifarnib (farnesyl transferase inhibitor), axitinib (multitargeted
resected. Resection of a pre-cancerous tumor should be considered tyrosine kinase inhibitor), matrix metalloprotease inhibitors have
the most efcient advance in pancreatic cancer management. generated disappointing results.
Most recently, a polychemotherapy regimen including 5-FU,
CHEMOTHERAPY leucovorin, oxaliplatin, and irinotecan was compared to gemc-
Chemotherapy may be used either in the palliative setting itabine as rst-line therapy in 342 metastatic pancreatic cancer
for metastatic or locally advanced unresectable tumors, or in patients, with good performance status, normal liver function and
resectable disease before (neoadjuvant) or after surgery (adjuvant age under 76. The median overall survival, which was the primary
treatment). endpoint, was 11.1 months in the FOLFIRINOX group as com-
pared with 6.8 months in the gemcitabine group [HR for death,
PALLIATIVE TREATMENT 0.57; 95% condence interval (CI), 0.450.73; P < 0.001]. Median
Gemcitabine, a nucleoside analog, became the reference treat- progression-free survival was 6.4 months in the FOLFIRINOX
ment for advanced pancreatic cancer after a randomized trial in group and 3.3 months in the gemcitabine group (HR for disease
which 126 chemotherapy-nave patients with advanced disease progression, 0.47; 95% CI, 0.370.59; P < 0.001). The objective
were randomized to receive either weekly gemcitabine or 5-FU response rate was 31.6% in the FOLFIRINOX group versus 9.4%
(Burris et al., 1997). No statistically signicant difference was in the gemcitabine group (P < 0.001). More adverse events were
found in the overall response rate between the two groups (5.4% noted in the FOLFIRINOX group with a higher incidence of
for gemcitabine and 0% for 5-FU), but the number of patient who grade 3 or 4 neutropenia, febrile neutropenia, thrombocytope-
experienced improved clinical response (improvement in pain or nia, diarrhea, and sensory neuropathy, as well as grade 2 alopecia.
performance status) was signicantly greater in the gemcitabine At 6 months, 31% of the patients in the FOLFIRINOX group
group than in the 5-FU group (23.8 versus 4.8%, P = 0.0022). had a denitive degradation of the quality of life versus 66% in

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 14
Iovanna et al. Pancreatic cancer

the gemcitabine group (HR, 0.47; 95% CI, 0.300.70; P < 0.001; chemoradiotherapy had a deleterious effect on survival (Ghaneh
Conroy et al., 2011). These results suggest that FOLFIRINOX and Neoptolemos, 2004). In 2007, Oettle et al. (2007) compared
should be considered as the recommended rst-line option for weekly gemcitabine (6 months) to observation after curative-
patients with metastatic pancreatic cancer and judged t to receive intent resection in 368 pancreatic cancer patients (CONKO-001
it (Performance status 01, normal, or nearly normal bilirubin trial). Median disease-free survival, the primary endpoint, was
level, age < 76). 13.4 months in the gemcitabine group and 6.9 months in the
In patients with non-metastatic locally advanced pancreatic control group (P < 0.001, log-rank; Oettle et al., 2007). Final
tumor (i.e., non-resectable), primary chemoradiation was the pre- results of this trial showed that this disease-free survival differ-
ferred treatment in the 90s. However, some patients received a ence translated into an overall survival advantage (Oettle et al.,
local treatment and were diagnosed with distant disease after 2007). Thus, both 5-FU- and gemcitabine-adjuvant chemother-
completion of chemoradiation. In addition, a French random- apy improve outcome in resected pancreatic cancer. A face to
ized trial comparing gemcitabine alone to chemoradiation (5- face comparative study between 5-FU and gemcitabine in this
FU + cisplatin) followed by gemcitabine, showed a lower over- setting revealed no difference in efcacy, but 5-FU appeared
all survival in the radiotherapy arm (Chauffert et al., 2008). more toxic (Neoptolemos et al., 2009). Therefore, gemcitabine
Thus, unresectable tumors are usually treated with short period is the recommended choice for adjuvant treatment, but 5-FU
chemotherapy followed by chemoradiation in patients with non- may be used as an alternative in case of intolerance. Again,
progressive disease. In addition, some of these patients may be after recent exciting results reported in the palliative setting,
secondarily brought to surgery. FOLFIRINOX will have to be evaluated as adjuvant treatment after
surgery.
NEOADJUVANT CHEMOTHERAPY
As judged from its impact on survival, neoadjuvant treatment RADIOTHERAPY
is not recommended for resectable pancreatic cancer. However, Radiotherapy is an efcient weapon against pancreatic cancer.
two uncontrolled phase II trials have evaluated neoadjuvant However, appropriate use/timing of radiotherapy was not precisely
gemcitabine-based chemotherapy alone (without radiation), with identied. Radiotherapy alone was not used in pancreatic cancer.
promising results. Palmer et al. (2007) randomized 50 patients Neoadjuvant chemoradiation was performed with uneven results
with potentially resectable pancreatic lesions to be treated with a in patients with resectable tumor. Several drugs were associated
short phase of weekly gemcitabine plus or minus cisplatin. Overall, to radiotherapy (5-FU, cisplatine, gemcitabine. . .) with similar
54% of patients underwent resection, 9 (38%) in the gemcitabine results. Patients who had had tumor resection after chemora-
arm, and 18 (70%) in the combination arm, with no increase diation showed histologic modications in the removed tumor.
in surgical complications. Twelve-month survival for the gemc- Moreover, sterilized specimens were sometimes observed. Thus,
itabine and combination groups was 42 and 62% (Palmer et al., chemoradiation is certainly efcient in a small proportion of
2007). In a prospective phase II study, Heinrich et al. (2008) tested patients but no factor could be identied at diagnosis to determine
neoadjuvant gemcitabinecisplatin biweekly, for four cycles in 28 which group of patient had to receive chemoradiation. To date,
patients with resectable adenocarcinoma of the pancreatic head. chemoradiation should not be recommended in resectable tumor.
Resection rate R0 was 80% and survival was increased (Heinrich Finally, in patients with locally advanced tumor, chemoradiation
et al., 2008). However, these studies were small-sized and further is used after inductive chemotherapy in patients with still non-
data are required before integrating preoperative chemotherapy metastatic disease. However, a small proportion of patients with
in the routine setting. Of course, because of the results reported locally advanced tumors are eligible for resection after completion
with FOLFIRINOX in advanced disease, this regimen should be of chemotherapy followed by chemoradiation.
imperatively evaluated in this strategy. Neoadjuvant chemoradia-
tion was also used in phase II studies but showed poor impact on NEW STRATEGIES FOR THE TREATMENT OF PANCREATIC
disease progression (Gillen et al., 2010). CANCERS
USING GENE THERAPY TO ENHANCE CANCER CHEMOTHERAPEUTIC
ADJUVANT TREATMENT EFFICACY
Till the mid-2000s, resection of pancreatic cancer was not rou- In recent years, gene therapy based on the knowledge of molecular
tinely associated with postoperative chemotherapy. In 2004, the pathogenesis has been actively developed as a novel therapeutic
nal results of a randomized trial (ESPAC-1) were reported, com- strategy to be used alone or in combination with conventional
paring surgery alone (n = 69) to chemotherapy alone (5-FU bolus chemotherapy in pancreatic cancer. Strategies for gene therapy in
plus folinic acid, 6 months; n = 75), radiochemotherapy (5-FU- pancreatic cancer include antisense and RNA interference strate-
based; n = 73), or both (n = 72). The estimated 5-year survival gies whereby the function of activated oncogenes (K-ras, H-ras,
rate was 10% among patients assigned to receive chemoradio- Notch, LSM1, etc.) is inhibited, and strategies to restore the func-
therapy and 20% among patients who did not receive chemora- tion of tumor suppressor genes (p53, p16/INK4a, p21/WAF1,
diotherapy (P = 0.05). The 5-year survival rate was 21% among DPC4/Smad4, etc.). These therapeutic strategies have shown their
patients who received chemotherapy and 8% among patients who promising effects on the inhibition of pancreatic cancer in vitro
did not receive chemotherapy (P = 0.009). These results indicate and in vivo (Bhattacharyya and Lemoine, 2006). In addition,
that adjuvant chemotherapy provided signicant survival bene- gene-directed pro-drug activation therapy is another gene ther-
t in patients with resected pancreatic cancer, whereas adjuvant apy system in which a gene that encodes an enzyme is delivered

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 15


Iovanna et al. Pancreatic cancer

to the tumor cell. Then, a pro-drug administered as chemother- considerable structural deviation from gemcitabine on one hand,
apy is metabolized by the enzyme to release cytotoxic drug at but also important structural diversity on the other hand. They
the site of the tumor. A recent report has shown that introduc- should be a good starting point to search for novel anticancer
tion of a fusion gene combining deoxycytidine kinase and uridine candidates against pancreatic cancer.
monophosphate kinase, which converts gemcitabine into its toxic All the synthesized triazole nucleosides types AD were rst
phosphorylated metabolite, sensitizes pancreatic cancer cells to assessed in vitro using MTT test to evaluate their antiprolif-
gemcitabine by reducing dramatically both in vitro cell viabil- erative activity on drug-sensitive (Capan2) and drug-resistant
ity and in vivo tumor volume (Vernejoul et al., 2006). Moreover, (MiaPaCa2) human pancreatic cancer cells. Among more than
the combinations of gemcitabine treatment with introduction of 600 compounds tested, dozens of compounds showed effective
IFN-, NK4, or p53 signicantly suppress the growth and the antiproliferative activity against drug-sensitive pancreatic cancer
metastasis of human pancreatic cancer cells, suggesting that the Capan2 cells, whereas a smaller amount of compounds emerged
efcacy of chemotherapy in pancreatic cancer can actually be with effective anticancer activity against drug-resistant pancre-
enhanced by gene therapy (Bhattacharyya and Lemoine, 2006; atic cancer MiaPaCa2 cells. Since we are particularly interested
Ogura et al., 2006). in identifying leads against drug-resistant pancreatic cancer, we
focused our attention on the most promising active compounds
PROMISING TRIAZOLE NUCLEOSIDE COMPOUNDS TO FIGHT named 1 and 2, which exhibited much better in vitro activity
DRUG-RESISTANT PANCREATIC CANCER than gemcitabine (Wan et al., 2009; Xia et al., 2009). Moreover,
We initiated, several years ago, a research program to develop novel both compounds 1 and 2 could efciently inhibit tumor growth
triazole nucleoside analogs with appended aromatic systems on in MiaPaCa2-xenograft nude mice after a 2-week treatment only,
the triazole base, in the view to searching for biologically active with no apparent adverse effect. Further studies revealed that the
nucleoside analogs as drug candidates against pancreatic cancer, induction of caspase-dependent apoptosis represented the main
particularly for drug-resistant pancreatic cancer. The rationale eason for the observed anticancer activity of compounds 1 and
for molecular design was based on the following considerations. 2. Although both compounds are originally designed as nucleo-
First, nucleoside analogs show important anticancer activity; sev- side analogs to interfere with DNA synthesis, neither compound
eral successful nucleoside drugs such as gemcitabine, cladribine, 1 nor compound 2 could signicantly inhibit DNA synthesis,
capecitabine, among others, are currently used as anticancer ther- contrary to gemcitabine. Gemcitabine has been also reported
apy (Galmarini et al., 2002). Second, triazole is an unnatural to down-regulate p8, a stress-associated protein with antiapop-
heterocycle, and nucleosides containing triazole as nucleobase totic properties. However, neither compound 1 nor compound
may be more resistant to nucleos(t)ide metabolizing enzymes and 2 could signicantly down-regulate p8 in a way similar similar
thus offer better in vivo stability. Third, triazole is considered as to gemcitabine. On the contrary, both of them rather upregu-
a universal base capable to form base-pairs with all ve natural late p8 expression. These ndings suggest that the mechanisms
nucleobases, it can therefore impart triazole nucleosides much of action of the active Triazole nucleosides compounds 1 and 2
better interaction with their biological receptors (Loakes, 2001). have mechanisms of action very different from those of gem-
Finally, appending aromatic systems to a triazole nucleobase may citabine. In fact, preliminary investigations demonstrated that
yield an expanded and enlarged conjugated with advantageous compound 2 could effectively down-regulate heat shock pro-
binding properties to the corresponding nucleosides for interac- tein 27 (Hsp27) at both mRNA and protein levels. Hsp27 is
tion with biological targets through stronger and more efcient an ATP-independent small molecular chaperone recently recog-
binding via larger aromatic systems. Based on all these ratio- nized as a novel target for anticancer therapy (Concannon et al.,
nales, we have designed various triazole nucleoside analogs with 2003; Arrigo et al., 2007). It plays an important role in the
appended aromatic groups on the triazole nucleobase. Their sugar resistance shown by patients with pancreatic cancer to gemc-
moiety is either ribose or acyclic glycogen and the triazole nucle- itabine and its function is to protect against drug toxicity (Mori-
obase is connected to the sugar moiety in such a way that either 3- Iwamoto et al., 2007). Recent studies with antisense oligonu-
or 5-aryltriazole nucleoside analogs can be obtained. The aromatic cleotides and siRNA molecules down-regulating Hsp27 demon-
moieties are introduced on the triazole ring via direct connec- strated potent caspase-dependent apoptotic anticancer activity
tion (type A), triple bond bridge (type B), triazolyl ring linker on prostate cancer (Rocchi et al., 2004, 2005). These antisense
(type C), or N -arylation (type D). Appending aromatic moieties oligonucleotides are currently in clinical trial for use in the treat-
directly on the tiazole ring provides biaryl motifs as nucleobase ment of several human cancers, including lung, breast, prostate,
in type A compounds. It is well known that biaryl motifs are bladder, and ovarian cancers. An additional study with IFN- also
very important in medicinal chemistry. Biaryl motifs as nucle- showed that down-regulation of Hsp27 via IFN- could gener-
obase in nucleoside analogs are of great interest in the view to ate effective anticancer activity in drug-resistant pancreatic cancer
exploring new nucleoside mimics. Connection of aromatic moi- cells, further validating the general utility and importance of
eties via triple bond bridge or triazolyl ring on the triazole ring down-regulating Hsp27 in treating drug-resistant cancers (Mori-
can offer enlarged conjugated aromatic systems as nucleobases, Iwamoto et al., 2009). This represents the rst small molecular
and may lead to better and stronger binding with biological tar- anticancer lead with such a novel mode of action and effective
gets. Amine linkage between aromatic moieties and triazole ring in anticancer consequence, opening a new avenue in the search for
type D is expected to provide some exibility to the correspond- novel anticancer drug candidates to ght drug-resistant pancreatic
ing nucleobases. Altogether, these compounds provide not only cancer.

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 16
Iovanna et al. Pancreatic cancer

CONCLUSION mutations, have been developed and they hold promise for future
The development and progression of pancreatic cancer are closely studies of the disease. The bioavailability of compounds such as
associated with the activation of oncogenes, the inactivation of antisense oligonucleotides and siRNAs in humans remains a big
tumor suppressor genes, the deregulation of EGFR, Akt, NFB, and hurdle, which will require further improvement of gene-delivery
their downstream signaling pathways. Although targeted thera- strategies.
pies for pancreatic cancer have yielded encouraging results in vitro The individualization of therapy for patients is possible if
and in animal models, these ndings have not been translated factors that predict treatment response, such as biological mark-
to improved outcomes in clinical trials. Reasons for this failure ers, can be determined accurately. These approaches are likely
might include an incomplete understanding of the biology of to comprise a mixture of targeted agents in combination with
pancreatic cancer, the selection of poor active agents, problems conventional chemotherapy and radiotherapy. For a clinically sig-
with trial design (such as inappropriate therapeutic end points or nicant effect to be achieved, treatment strategies should either be
patient selection) and the rapidity with which agents move into in the form of a horizontal approach, in which several oncogenic
randomized, controlled trials without the extensive early testing pathways are inhibited, or a vertical approach, whereby multi-
necessary to optimize treatment regimens. Furthermore, preclini- ple levels of a major pathway are targeted. Combined therapies,
cal studies performed on mouse models do not always recapitulate together with improved diagnostic tools and predictive markers,
the human condition, a particularly important limitation with are ultimately hoped to improve the bleak outlook of patients
human pancreatic cancer xenografts in immunodecient mice. diagnosed with pancreatic cancer.
Despite these setbacks, lessons have been learnt, and our col-
lective effort has generated a substantial platform of knowledge ACKNOWLEDGMENTS
from which further work could spring. Genetically engineered This work was supported by grants from INSERM, Ligue Contre
immunocompetent mice, such as those with K-RAS or TP53 le Cancer, and INCA.

REFERENCES LaCroix, A., Zheng, W., Albanes, D., (HspB5) as therapeutic targets. FEBS J., Tsai, M. M., Zhu, M., Vonderfecht,
Abdelrahim, M., Newman, K., Van- Bamlet, W., Berg, C. D., Berrino, F., Lett. 581, 36653674. S., Baserga, R., Kendall, R., Radinsky,
derlaag, K., Samudio, I., and Safe, Bingham, S., Buring, J. E., Bracci, Asano, T., Yao, Y., Zhu, J., Li, D., R., and Calzone, F. J. (2009). AMG
S. (2006). 3,3 -Diindolylmethane P. M., Canzian, F., Clavel-Chapelon, Abbruzzese, J. L., and Reddy, S. A. 479, a fully human anti-insulin-like
(DIM) and its derivatives induce F., Clipp, S., Cotterchio, M., de (2004). The PI 3-kinase/Akt sig- growth factor receptor type I mono-
apoptosis in pancreatic cancer cells Andrade, M., Duell, E. J., Fox, J. naling pathway is activated due to clonal antibody, inhibits the growth
through endoplasmic reticulum W. Jr., Gallinger, S., Gaziano, J. M., aberrant Pten expression and tar- and survival of pancreatic carci-
stress-dependent upregulation of Giovannucci, E. L., Goggins, M., gets transcription factors NF-B and noma cells. Mol. Cancer Ther. 8,
DR5. Carcinogenesis 27, 717728. Gonzlez, C. A., Hallmans, G., Han- c-Myc in pancreatic cancer cells. 10951105.
Abe, N., Watanabe, T., Masaki, T., Mori, kinson, S. E., Hassan, M., Holly, E. Oncogene 23, 85718580. Berger, S. L. (2002). Histone modica-
T., Sugiyama, M., Uchimura, H., A., Hunter, D. J., Hutchinson, A., Baker, C. H., Trevino, J. G., Summy, tions in transcriptional regulation.
Fujioka, Y., Chiappetta, G., Fusco, Jackson, R., Jacobs, K. B., Jenab, J. M., Zhang, F., Caron, A., Nesbit, Curr. Opin. Genet. Dev. 12, 142148.
A., and Atomi, Y. (2000). Pancreatic M., Kaaks, R., Klein, A. P., Kooper- M., Gallick, G. E., and Fidler, I. J. Berlin, J. D., Catalano, P., Thomas, J.
duct cell carcinomas express high berg, C., Kurtz, R. C., Li, D., Lynch, (2006). Inhibition of PDGFR phos- P., Kugler, J. W., Haller, D. G., and
levels of high mobility group I(Y) S. M., Mandelson, M., McWilliams, phorylation and Src and Akt activ- Benson, A. B. III. (2002). Phase III
proteins. Cancer Res. 60, 31173122. R. R., Mendelsohn, J. B., Michaud, ity by GN963 leads to therapy of study of gemcitabine in combination
Abraham, S. C., Wu, T. T., Klim- D. S., Olson, S. H., Overvad, K., human pancreatic cancer growing with uorouracil versus gemcitabine
stra, D. S., Finn, L. S., Lee, J. H., Patel, A. V., Peeters, P. H., Rajkovic, orthotopically in nude mice. Int. J. alone in patients with advanced pan-
Yeo, C. J., Cameron, J. L., and A., Riboli, E., Risch, H. A., Shu, X. Oncol. 29, 125138. creatic carcinoma: Eastern Coopera-
Hruban, R. H. (2001). Distinc- O., Thomas, G., Tobias, G. S., Tri- Banerjee, S., Zhang, Y., Ali, S., Bhuiyan, tive Oncology Group Trial E2297. J.
tive molecular genetic alterations in chopoulos, D., Van Den Eeden, S. M., Wang, Z., Chiao, P. J., Philip, Clin. Oncol. 20, 32703275.
sporadic and familial adenomatous K., Virtamo, J., Wactawski-Wende, P. A., Abbruzzese, J., and Sarkar, F. Berman, D. M., Karhadkar, S. S., Maitra,
polyposis-associated pancreatoblas- J., Wolpin, B. M., Yu, H., Yu, K., H. (2005). Molecular evidence for A., Montes De Oca, R., Gerstenblith,
tomas: frequent alterations in the Zeleniuch-Jacquotte, A., Chanock, S. increased antitumor activity of gem- M. R., Briggs, K., Parker, A. R., Shi-
APC/-catenin pathway and chro- J., Hartge, P., and Hoover, R. N. citabine by genistein in vitro and mada, Y., Eshleman, J. R., Watkins,
mosome 11p. Am. J. Pathol. 159, (2009). Genome-wide association in vivo using an orthotopic model D. N., and Beachy, P. A. (2003).
16191627. study identies variants in the ABO of pancreatic cancer. Cancer Res. 65, Widespread requirement for Hedge-
Aird, I., Bentall, H. H., and Roberts, J. A. locus associated with susceptibility 90649072. hog ligand stimulation in growth of
(1953). A relationship between can- to pancreatic cancer. Nat. Genet. 41, Banville, N., Geraghty, R., Fox, E., Leahy, digestive tract tumours. Nature 425,
cer of stomach and the ABO blood 986990. D. T., Green, A., Keegan, D., Geoghe- 846851.
groups. Br. Med. J. 1, 799801. Andea, A., Sarkar, F., and Adsay, V. gan, J., ODonoghue, D., Hyland, J., Bernhardt, S. L., Gjertsen, M. K., Trach-
Almoguera, C., Shibata, D., Forrester, K., N. (2003). Clinicopathological cor- and Sheahan, K. (2006). Medullary sel, S., Moller, M., Eriksen, J. A., Meo,
Martin, J., Arnheim, N., and Peru- relates of pancreatic intraepithelial carcinoma of the pancreas in a M., Buanes, T., and Gaudernack, G.
cho, M. (1988). Most human carci- neoplasia: a comparative analysis of man with hereditary nonpolyposis (2006). Telomerase peptide vaccina-
nomas of the exocrine pancreas con- 82 cases with and 152 cases without colorectal cancer due to a mutation tion of patients with non-resectable
tain mutant c-K-ras genes. Cell 53, pancreatic ductal adenocarcinoma. of the MSH2 mismatch repair gene. pancreatic cancer: a dose escalating
549554. Mod. Pathol. 16, 9961006. Hum. Pathol. 37, 14981502. phase I/II study. Br. J. Cancer 95,
Amundadottir, L., Kraft, P., Stolzenberg- Arrigo, A. P., Simon, S., Gibert, B., Bardeesy, N., and DePinho, R. A. (2002). 14741482.
Solomon, R. Z., Fuchs, C. S., Kretz-Remy, C., Nivon, M., Czekalla, Pancreatic cancer biology and genet- Bhattacharyya, M., and Lemoine, N. R.
Petersen, G. M., Arslan, A. A., A., Guillet, D., Moulin, M., Diaz- ics. Nat. Rev. Cancer 2, 897909. (2006). Gene therapy developments
Bueno-de-Mesquita, H. B., Gross, Latoud, C., and Vicart, P. (2007). Beltran, P. J., Mitchell, P., Chung, Y. A., for pancreatic cancer. Best Pract. Res.
M., Helzlsouer, K., Jacobs, E. J., Hsp27 (HspB1) and B-crystallin Cajulis, E., Lu, J., Belmontes, B., Ho, Clin. Gastroenterol. 20, 285298.

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 17


Iovanna et al. Pancreatic cancer

Biliran, H. Jr., Wang, Y., Banerjee, S., advanced pancreas cancer: a ran- III study of the Gruppo Oncologia Tudur-Smith, C., West, J., Falk,
Xu, H., Heng, H., Thakur, A., Bol- domized trial. J. Clin. Oncol. 15, dellItalia Meridionale. Cancer 94, S., Crellin, A., Adab, F., Thomp-
lig, A., Sarkar, F. H., and Liao, J. 24032413. 902910. son, J., Leonard, P., Ostrowski, J.,
D. (2005). Overexpression of cyclin Campbell, P. J., Yachida, S., Mudie, L. Colucci, G., Labianca, R., Di Costanzo, Eatock, M., Scheithauer, W., Her-
D1 promotes tumor cell growth J., Stephens, P. J., Pleasance, E. D., F., Gebbia, V., Carteni, G., Mas- rmann, R., and Neoptolemos, J.
and confers resistance to cisplatin- Stebbings, L. A., Morsberger, L. A., sidda, B., Dapretto, E., Manzione, P. (2009). Phase III randomized
mediated apoptosis in an elastase- Latimer, C., McLaren, S., Lin, M. L., L., Piazza, E., Sannicolo, M., Cia- comparison of gemcitabine ver-
myc transgene-expressing pancre- McBride, D. J., Varela, I., Nik-Zainal, parrone, M., Cavanna, L., Giuliani, sus gemcitabine plus capecitabine
atic tumor cell line. Clin. Cancer Res. S. A., Leroy, C., Jia, M., Menzies, A., F., Maiello, E., Testa, A., Pederzoli, in patients with advanced pan-
11, 60756086. Butler, A. P., Teague, J. W., Grifn, C. P., Falconi, M., Gallo, C., Di Maio, creatic cancer. J. Clin. Oncol. 27,
Bonafoux, D., and Lee, W. C. (2009). A., Burton, J., Swerdlow, H., Quail, M., Perrone, F., Gruppo Oncologico 55135518.
Strategies for TGF- modulation: M. A., Stratton, M. R., Iacobuzio- Italia Meridionale (GOIM), Gruppo Dhillon, N., Aggarwal, B. B., Newman,
a review of recent patents. Expert Donahue, C., and Futreal, P. A. Italiano per lo Studio dei Carcinomi R. A., Wolff, R. A., Kunnumakkara,
Opin. Ther. Pat. 19, 17591769. (2010). The patterns and dynamics dellApparato Digerente (GISCAD), A. B., Abbruzzese, J. L., Ng, C.
Bramhall, S. R., Rosemurgy, A., Brown, of genomic instability in metasta- and Gruppo Oncologico Italiano di S., Badmaev, V., and Kurzrock, R.
P. D., Bowry, C., and Buckels, J. A. tic pancreatic cancer. Nature 467, Ricerca Clinica (GOIRC). (2010). (2008). Phase II trial of curcumin
(2001). Marimastat as rst-line ther- 11091113. Randomized phase III trial of gemc- in patients with advanced pancre-
apy for patients with unresectable Cao, R., Wang, L., Wang, H., Xia, L., itabine plus cisplatin compared with atic cancer. Clin. Cancer Res. 14,
pancreatic cancer: a randomized Erdjument-Bromage, H., Tempst, P., single-agent gemcitabine as rst-line 44914499.
trial. J. Clin. Oncol. 19, 34473455. Jones, R. S., and Zhang, Y. (2002). treatment of patients with advanced Diergaarde, B., Brand, R., Lamb, J.,
Bramhall, S. R., Schulz, J., Nemu- Role of histone H3 lysine 27 methy- pancreatic cancer: the GIP-1 study. J. Cheong, S. Y., Stello, K., Barmada, M.
naitis, J., Brown, P. D., Baillet, lation in polycomb-group silencing. Clin. Oncol. 28, 16451651. M., Feingold, E., and Whitcomb, D.
M., and Buckels, J. A. (2002). Science 298, 10391043. Concannon, C. G., Gorman, A. M., C. (2010). Pooling-based genome-
A double-blind placebo-controlled, Caplin, M., Savage, K., Khan, K., and Samali, A. (2003). On the role wide association study implicates -
randomised study comparing gem- Brett, B., Rode, J., Varro, A., and of Hsp27 in regulating apoptosis. glutamyltransferase 1 (GGT1) gene
citabine and marimastat with gem- Dhillon, A. (2000). Expression and Apoptosis 8, 6170. in pancreatic carcinogenesis. Pancre-
citabine and placebo as rst line processing of gastrin in pancreatic Conroy, T., Desseigne, F., Ychou, atology 10, 194200.
therapy in patients with advanced adenocarcinoma. Br. J. Surg. 87, M., Bouche, O., Guimbaud, R., Ding, X. Z., and Adrian, T. E. (2002).
pancreatic cancer. Br. J. Cancer 87, 10351040. Becouarn, Y., Adenis, A., Raoul, Resveratrol inhibits proliferation
161167. Chau, I., Cunningham, D., Russell, C., J. L., Gourgou-Bourgade, S., de and induces apoptosis in human
Brat, D. J., Lillemoe, K. D., Yeo, C. Norman, A. R., Kurzawinski, T., la Fouchardiere, C., Bennouna, J., pancreatic cancer cells. Pancreas 25,
J., Wareld, P. B., and Hruban, R. Harper, P., Harrison, P., Middleton, Bachet, J. B., Khemissa-Akouz, F., e71e76.
H. (1998). Progression of pancreatic G., Daniels, F., Hickish, T., Pren- Pr-Verg, D., Delbaldo, C., Asse- Donadelli, M., Costanzo, C., Beghelli,
intraductal neoplasias to inltrat- deville, J., Ross, P. J., Theis, B., nat, E., Chauffert, B., Michel, P., S., Scupoli, M. T., Dandrea, M.,
ing adenocarcinoma of the pancreas. Hull, R., Walker, M., Shankley, N., Montoto-Grillot, C., Ducreux, M., Bonora, A., Piacentini, P., Budil-
Am. J. Surg. Pathol. 22, 163169. Kalindjian, B., Murray, G., Gillbanks, Groupe Tumeurs Digestives of Uni- lon, A., Caraglia, M., Scarpa, A.,
Brockie, E., Anand, A., and Albores- A., and Black, J. (2006). Gastrazole cancer, and PRODIGE Intergroup. and Palmieri, M. (2007). Synergistic
Saavedra, J. (1998). Progression (JB95008), a novel CCK2/gastrin (2011). FOLFIRINOX versus gem- inhibition of pancreatic adenocarci-
of atypical ductal hyperpla- receptor antagonist, in the treat- citabine for metastatic pancreatic noma cell growth by trichostatin A
sia/carcinoma in situ of the pancreas ment of advanced pancreatic can- cancer. N. Engl. J. Med. 364, and gemcitabine. Biochim. Biophys.
to invasive adenocarcinoma. Ann. cer: results from two randomised 18171825. Acta 1773, 10951106.
Diagn. Pathol. 2, 286292. controlled trials. Br. J. Cancer 94, Corcoran, R. B., Contino, G., Desh- Dong, J. T., and Chen, C. (2009). Essen-
Bruserud, O., Stapnes, C., Ersvaer, 11071115. pande, V., Tzatsos, A., Conrad, C., tial role of KLF5 transcription fac-
E., Gjertsen, B. T., and Rynin- Chauffert, B., Mornex, F., Bonnetain, Benes, C. H., Levy, D. E., Settleman, tor in cell proliferation and differ-
gen, A. (2007). Histone deacetylase F., Rougier, P., Mariette, C., Bouche, J., Engelman, J. A., and Bardeesy, N. entiation and its implications for
inhibitors in cancer treatment: a O., Bosset, J. F., Aparicio, T., (2011). STAT3 plays a critical role in human diseases. Cell. Mol. Life Sci.
review of the clinical toxicity and the Mineur, L., Azzedine, A., Ham- KRAS-induced pancreatic tumori- 66, 26912706.
modulation of gene expression in mel, P., Butel, J., Stremsdoerfer, genesis. Cancer Res. 71, 50205029. Earl, J., Yan, L., Vitone, L. J., Risk,
cancer cell. Curr. Pharm. Biotechnol. N., Maingon, P., and Bedenne, L. Coughlin, S. S., Calle, E. E., Patel, A. V., J., Kemp, S. J., McFaul, C., Neop-
8, 388400. (2008). Phase III trial comparing and Thun, M. J. (2000). Predictors of tolemos, J. P., Greenhalf, W., Euro-
Buchler, P., Gazdhar, A., Schubert, M., intensive induction chemoradio- pancreatic cancer mortality among a pean Registry of Hereditary Pancre-
Giese, N., Reber, H. A., Hines, O. therapy (60 Gy, infusional 5-FU and large cohort of United States adults. atitis and Familial Pancreatic Cancer,
J., Giese, T., Ceyhan, G. O., Muller, intermittent cisplatin) followed by Cancer Causes Control 11, 915923. Kress, R., Sina-Frey, M., Hahn, S. A.,
M., Buchler, M. W., and Friess, maintenance gemcitabine with gem- Cowgill, S. M., and Muscarella, P. Rieder, H., Bartsch, D. K., and Ger-
H. (2005). The Notch signaling citabine alone for locally advanced (2003). The genetics of pancreatic man National Case Collection for
pathway is related to neurovascu- unresectable pancreatic cancer. cancer. Am. J. Surg. 186, 279286. Familial Pancreatic Cancer. (2006).
lar progression of pancreatic cancer. Denitive results of the 2000-01 Cress, W. D., and Seto, E. (2000). Evaluation of the 4q32-34 locus in
Ann. Surg. 242, 791800; discussion FFCD/SFRO study. Ann. Oncol. 19, Histone deacetylases, transcriptional European familial pancreatic cancer.
800801. 15921599. control, and cancer. J. Cell. Physiol. Cancer Epidemiol. Biomarkers Prev.
Burris, H. A. III, Moore, M. J., Ander- Colucci, G., Giuliani, F., Gebbia, V., 184, 116. 15, 19481955.
sen, J., Green, M. R., Rothenberg, Biglietto, M., Rabitti, P., Uomo, Cubilla, A. L., and Fitzgerald, P. J. El-Rayes, B. F., Ali, S., Sarkar, F.
M. L., Modiano, M. R., Cripps, M. G., Cigolari, S., Testa, A., Maiello, (1976). Morphological lesions asso- H., and Philip, P. A. (2004).
C., Portenoy, R. K., Storniolo, A. M., E., and Lopez, M. (2002). Gem- ciated with human primary inva- Cyclooxygenase-2-dependent and -
Tarassoff, P., Nelson, R., Dorr, F. A., citabine alone or with cisplatin sive nonendocrine pancreas cancer. independent effects of celecoxib in
Stephens, C. D., and Von Hoff, D. for the treatment of patients Cancer Res. 36, 26902698. pancreatic cancer cell lines. Mol.
D. (1997). Improvements in survival with locally advanced and/or Cunningham, D., Chau, I., Stocken, D. Cancer Ther. 3, 14211426.
and clinical benet with gemcitabine metastatic pancreatic carcinoma: D., Valle, J. W., Smith, D., Stew- Esteller, M. (2008). Epigenetics in can-
as rst-line therapy for patients with a prospective, randomized phase ard, W., Harper, P. G., Dunn, J., cer. N. Engl. J. Med. 358, 11481159.

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 18
Iovanna et al. Pancreatic cancer

Fahy, B. N., Schlieman, M. G., Viru- inhibitor suppressing an expression expression proling of multiple his- Habbe, N., Koorstra, J. B., Mendell,
dachalam, S., and Bold, R. J. of integrin 2 subunit on endothe- tone deacetylase (HDAC) inhibitors: J. T., Offerhaus, G. J., Ryu, J. K.,
(2004). Inhibition of AKT abro- lium. Cancer Res. 62, 61166123. dening a common gene set pro- Feldmann, G., Mullendore, M. E.,
gates chemotherapy-induced NF- Furukawa, T. (2007). Molecular genetics duced by HDAC inhibition in T24 Goggins, M. G., Hong, S. M., and
B survival mechanisms: implica- of intraductal papillary-mucinous and MDA carcinoma cell lines. Mol. Maitra, A. (2009). MicroRNA miR-
tions for therapy in pancreatic neoplasms of the pancreas. J. Hepa- Cancer Ther. 2, 151163. 155 is a biomarker of early pancre-
cancer. J. Am. Coll. Surg. 198, tobiliary Pancreat. Surg. 14, 233237. Goggins, M., Offerhaus, G. J., Hilgers, atic neoplasia. Cancer Biol. Ther. 8,
591599. Furukawa, T., Duguid, W. P., Kobari, W., Grifn, C. A., Shekher, M., Tang, 340346.
Fayard, E., Schoonjans, K., Annicotte, M., Matsuno, S., and Tsao, M. D., Sohn, T. A., Yeo, C. J., Kern, S. Hakam, A., Fang, Q., Karl, R., and Cop-
J. S., and Auwerx, J. (2003). Liver S. (1995). Hepatocyte growth fac- E., and Hruban, R. H. (1998a). Pan- pola, D. (2003). Coexpression of
receptor homolog 1 controls the tor and Met receptor expression creatic adenocarcinomas with DNA IGF-1R and c-Src proteins in human
expression of carboxyl ester lipase. in human pancreatic carcinogenesis. replication errors (RER+) are asso- pancreatic ductal adenocarcinoma.
J. Biol. Chem. 278, 3572535731. Am. J. Pathol. 147, 889895. ciated with wild-type K-ras and Dig. Dis. Sci. 48, 19721978.
Fearon, E. R., and Vogelstein, B. (1990). Furuyama, K., Doi, R., Mori, T., Toy- characteristic histopathology. Poor Heinemann, V., Boeck, S., Hinke, A.,
A genetic model for colorectal oda, E., Ito, D., Kami, K., Koizumi, differentiation, a syncytial growth Labianca, R., and Louvet, C. (2008).
tumorigenesis. Cell 61, 759767. M., Kida, A., Kawaguchi, Y., and pattern, and pushing borders sug- Meta-analysis of randomized tri-
Feldmann, G., Dhara, S., Fendrich, V., Fujimoto, K. (2006). Clinical signif- gest RER+. Am. J. Pathol. 152, als: evaluation of benet from
Bedja, D., Beaty, R., Mullendore, M., icance of focal adhesion kinase in 15011507. gemcitabine-based combination
Karikari, C., Alvarez, H., Iacobuzio- resectable pancreatic cancer. World Goggins, M., Shekher, M., Turnacioglu, chemotherapy applied in advanced
Donahue, C., Jimeno, A., Gabriel- J. Surg. 30, 219226. K., Yeo, C. J., Hruban, R. H., and pancreatic cancer. BMC Cancer 8,
son, K. L., Matsui, W., and Maitra, Galmarini, C. M., Mackey, J. R., and Kern, S. E. (1998b). Genetic alter- 82. doi:10.1186/1471-2407-8-82
A. (2007). Blockade of hedgehog Dumontet, C. (2002). Nucleoside ations of the transforming growth Heinemann, V., Quietzsch, D., Gieseler,
signaling inhibits pancreatic can- analogues and nucleobases in can- factor receptor genes in pancre- F., Gonnermann, M., Schonekas, H.,
cer invasion and metastases: a new cer treatment. Lancet Oncol. 3, atic and biliary adenocarcinomas. Rost, A., Neuhaus, H., Haag, C.,
paradigm for combination therapy 415424. Cancer Res. 58, 53295332. Clemens, M., Heinrich, B., Vehling-
in solid cancers. Cancer Res. 67, Garcea, G., Neal, C. P., Pattenden, C. Goldstein, A. M., Fraser, M. C., Struew- Kaiser, U., Fuchs, M., Fleckenstein,
21872196. J., Steward, W. P., and Berry, D. P. ing, J. P., Hussussian, C. J., Ranade, D., Gesierich, W., Uthgenannt, D.,
Fernandez-Zapico, M. E., Gonzalez-Paz, (2005). Molecular prognostic mark- K., Zametkin, D. P., Fontaine, L. S., Einsele, H., Holstege, A., Hinke, A.,
N. C., Weiss, E., Savoy, D. N., Molina, ers in pancreatic cancer: a systematic Organic, S. M., Dracopoli, N. C., Schalhorn, A., and Wilkowski, R.
J. R., Fonseca, R., Smyrk, T. C., Chari, review. Eur. J. Cancer 41, 22132236. Clark, W. H. Jr., and Tucker, M. A. (2006). Randomized phase III trial
S. T., Urrutia, R., and Billadeau, D. D. Ghaneh, P., and Neoptolemos, J. P. (1995). Increased risk of pancreatic of gemcitabine plus cisplatin com-
(2005). Ectopic expression of VAV1 (2004). Conclusions from the Euro- cancer in melanoma-prone kindreds pared with gemcitabine alone in
reveals an unexpected role in pan- pean Study Group for Pancreatic with p16INK4 mutations. N. Engl. J. advanced pancreatic cancer. J. Clin.
creatic cancer tumorigenesis. Cancer Cancer adjuvant trial of chemora- Med. 333, 970974. Oncol. 24, 39463952.
Cell 7, 3949. diotherapy and chemotherapy for Gonalves, A., Viret, F., Franois, E., Heinrich, S., Pestalozzi, B. C., Schafer,
Friess, H., Langrehr, J. M., Oettle, H., pancreatic cancer. Surg. Oncol. Clin. Dahan, L., Perrier, H., Lamy, R., Re, M., Weber, A., Bauerfeind, P., Knuth,
Raedle, J., Niedergethmann, M., Dit- N. Am. 13, 567587. D., Largillier, R., Gasmi, M., Tchik- A., and Clavien, P. A. (2008).
trich, C., Hossfeld, D. K., Stoger, Giardiello, F. M., Brensinger, J. D., navorian, X., Turrini, O., Moureau- Prospective phase II trial of neoad-
H., Neyns, B., Herzog, P., Piedbois, Tersmette, A. C., Goodman, S. N., Zabotto, L., Delpero, J., and Viens, juvant chemotherapy with gemc-
P., Dobrowolski, F., Scheithauer, W., Petersen, G. M., Booker, S. V., Cruz- P. (2011). BAYPAN study: a double- itabine and cisplatin for resectable
Hawkins, R., Katz, F., Balcke, P., Ver- Correa, M., and Offerhaus, J. A. blind, phase III randomized trial of adenocarcinoma of the pancre-
morken, J., van Belle, S., Davidson, (2000). Very high risk of cancer gemcitabine plus sorafenib versus atic head. J. Clin. Oncol. 26,
N., Esteve, A. A., Castellano, D., Kle- in familial Peutz-Jeghers syndrome. gemcitabine plus placebo in patients 25262531.
eff, J., Tempia-Caliera, A. A., Kovar, Gastroenterology 119, 14471453. with advanced pancreatic cancer. J. Hermann, P. C., Huber, S. L., Her-
A., and Nippgen, J. (2006). A ran- Gillen, S., Schuster, T., Meyer Zum Clin. Oncol. 29(suppl.), abstr 4028. rler, T., Aicher, A., Ellwart, J.
domized multi-center phase II trial Buschenfelde, C., Friess, H., Grifths, E. A., and Gore, S. D. (2008). W., Guba, M., Bruns, C. J., and
of the angiogenesis inhibitor cilen- and Kleeff, J. (2010). Preop- DNA methyltransferase and histone Heeschen, C. (2007). Distinct pop-
gitide (EMD 121974) and gemc- erative/neoadjuvant therapy in deacetylase inhibitors in the treat- ulations of cancer stem cells deter-
itabine compared with gemcitabine pancreatic cancer: a systematic ment of myelodysplastic syndromes. mine tumor growth and metasta-
alone in advanced unresectable pan- review and meta-analysis of Semin. Hematol. 45, 2330. tic activity in human pancre-
creatic cancer. BMC Cancer 6, 285. response and resection percentages. Guerra, C., Collado, M., Navas, C., atic cancer. Cell Stem Cell 1,
doi:10.1186/1471-2407-6-285 PLoS Med. 7, e1000267. doi: 10.1371/ Schuhmacher, A. J., Hernandez- 313323.
Fujioka, S., Sclabas, G. M., Schmidt, journal.pmed.1000267 Porras, I., Canamero, M., Rodriguez- Herrmann, R., Bodoky, G., Ruhstaller,
C., Frederick, W. A., Dong, Q. G., Gjertsen, M. K., Buanes, T., Rosseland, Justo, M., Serrano, M., and Bar- T., Glimelius, B., Bajetta, E., Schuller,
Abbruzzese, J. L., Evans, D. B., Baker, A. R., Bakka, A., Gladhaug, I., Sor- bacid, M. (2011). Pancreatitis- J., Saletti, P., Bauer, J., Figer, A.,
C., and Chiao, P. J. (2003). Function eide, O., Eriksen, J. A., Moller, M., induced inammation contributes Pestalozzi, B., Khne, C. H., Min-
of nuclear factor B in pancreatic Baksaas, I., Lothe, R. A., Saeterdal, to pancreatic cancer by inhibiting grone, W., Stemmer, S. M., Tmas,
cancer metastasis. Clin. Cancer Res. I., and Gaudernack, G. (2001). oncogene-induced senescence. Can- K., Kornek, G. V., Koeberle, D.,
9, 346354. Intradermal ras peptide vaccina- cer Cell 19, 728739. Cina, S., Bernhard, J., Dietrich, D.,
Funahashi, Y., Sugi, N. H., Semba, tion with granulocyte-macrophage Guerra, C., Schuhmacher, A. J., Scheithauer, W., Swiss Group for
T., Yamamoto, Y., Hamaoka, S., colony-stimulating factor as adju- Canamero, M., Grippo, P. J., Verda- Clinical Cancer Research, and Cen-
Tsukahara-Tamai, N., Ozawa, Y., vant: clinical and immunological guer, L., Perez-Gallego, L., Dubus, tral European Cooperative Oncol-
Tsuruoka, A., Nara, K., Takahashi, responses in patients with pancre- P., Sandgren, E. P., and Barbacid, ogy Group. (2007). Gemcitabine
K., Okabe, T., Kamata, J., Owa, T., atic adenocarcinoma. Int. J. Cancer M. (2007). Chronic pancreatitis is plus capecitabine compared with
Ueda, N., Haneda, T., Yonaga, M., 92, 441450. essential for induction of pancreatic gemcitabine alone in advanced pan-
Yoshimatsu, K., and Wakabayashi, Glaser, K. B., Staver, M. J., Waring, ductal adenocarcinoma by K-Ras creatic cancer: a randomized, mul-
T. (2002). Sulfonamide derivative, J. F., Stender, J., Ulrich, R. G., oncogenes in adult mice. Cancer ticenter, phase III trial of the Swiss
E7820, is a unique angiogenesis and Davidsen, S. K. (2003). Gene Cell 11, 291302. Group for Clinical Cancer Research

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 19


Iovanna et al. Pancreatic cancer

and the Central European Coopera- Jang, M., Cai, L., Udeani, G. O., Slow- Cancer Chemother. Pharmacol. 61, Li, Y., Ellis, K. L., Ali, S., El-Rayes, B.
tive Oncology Group. J. Clin. Oncol. ing, K. V., Thomas, C. F., Beecher, 883892. F., Nedeljkovic-Kurepa, A., Kucuk,
25, 22122217. C. W., Fong, H. H., Farnsworth, Khan, N., Afaq, F., Saleem, M., O., Philip, P. A., and Sarkar,
Hildmann, C., Riester, D., and Schwien- N. R., Kinghorn, A. D., Mehta, R. Ahmad, N., and Mukhtar, H. F. H. (2004b). Apoptosis-inducing
horst, A. (2007). Histone deacety- G., Moon, R. C., and Pezzuto, J. (2006). Targeting multiple signal- effect of chemotherapeutic agents is
lases an important class of cellular M. (1997). Cancer chemopreven- ing pathways by green tea polyphe- potentiated by soy isoavone genis-
regulators with a variety of func- tive activity of resveratrol, a natural nol ()-epigallocatechin-3-gallate. tein, a natural inhibitor of NF-B in
tions. Appl. Microbiol. Biotechnol. 75, product derived from grapes. Science Cancer Res. 66, 25002505. BxPC-3 pancreatic cancer cell line.
487497. 275, 218220. Kindler, H. L., Niedzwiecki, D., Hol- Pancreas 28, e90e95.
Hiyama, E., Kodama, T., Shinbara, K., Jemal, A., Siegel, R., Ward, E., Murray, lis, D., Sutherland, S., Schrag, D., Li, Y., Ahmed, F., Ali, S., Philip, P.
Iwao, T., Itoh, M., Hiyama, K., Shay, T., Xu, J., Smigal, C., and Thun, M. Hurwitz, H., Innocenti, F., Mulc- A., Kucuk, O., and Sarkar, F. H.
J. W., Matsuura, Y., and Yokoyama, J. (2006). Cancer statistics, 2006. CA ahy, M. F., OReilly, E., Woz- (2005). Inactivation of nuclear fac-
T. (1997). Telomerase activity is Cancer J. Clin. 56, 106130. niak, T. F., Picus, J., Bhargava, tor B by soy isoavone genistein
detected in pancreatic cancer but not Jiang, J., Lee, E. J., Gusev, Y., and P., Mayer, R. J., Schilsky, R. L., contributes to increased apoptosis
in benign tumors. Cancer Res. 57, Schmittgen, T. D. (2005). Real-time and Goldberg, R. M. (2010). Gem- induced by chemotherapeutic agents
326331. expression proling of microRNA citabine plus bevacizumab com- in human cancer cells. Cancer Res.
Hruban, R. H., Adsay, N. V., Albores- precursors in human cancer pared with gemcitabine plus placebo 65, 69346942.
Saavedra, J., Compton, C., Garrett, E. cell lines. Nucleic Acids Res. 33, in patients with advanced pancre- Li, Y., Bhuiyan, M., Vaitkevicius, V. K.,
S., Goodman, S. N., Kern, S. E., Klim- 53945403. atic cancer: phase III trial of the and Sarkar, F. H. (1998). Molecu-
stra, D. S., Kloppel, G., Longnecker, Jimeno, A., Rubio-Viqueira, B., Cancer and Leukemia Group B lar analysis of the p53 gene in pan-
D. S., Lttges, J., and Offerhaus, G. Amador, M. L., Grunwald, V., (CALGB 80303). J. Clin. Oncol. 28, creatic adenocarcinoma. Diagn. Mol.
J. (2001). Pancreatic intraepithelial Maitra, A., Iacobuzio-Donahue, 36173622. Pathol. 7, 49.
neoplasia: a new nomenclature and C., and Hidalgo, M. (2007). Dual Kornmann, M., Arber, N., and Korc, Li,Y., Kucuk, O., Hussain, M.,Abrams, J.,
classication system for pancreatic mitogen-activated protein kinase M. (1998). Inhibition of basal and Cher, M. L., and Sarkar, F. H. (2006).
duct lesions. Am. J. Surg. Pathol. 25, and epidermal growth factor mitogen-stimulated pancreatic can- Antitumor and antimetastatic activ-
579586. receptor inhibition in biliary and cer cell growth by cyclin D1 anti- ities of docetaxel are enhanced
Hruban, R. H., Goggins, M., Parsons, J., pancreatic cancer. Mol. Cancer Ther. sense is associated with loss of by genistein through regulation of
and Kern, S. E. (2000). Progression 6, 10791088. tumorigenicity and potentiation of osteoprotegerin/receptor activator
model for pancreatic cancer. Clin. Jin, H., Yang, R., Zheng, Z., Romero, cytotoxicity to cisplatinum. J. Clin. of nuclear factor-B (RANK)/RANK
Cancer Res. 6, 29692972. M., Ross, J., Bou-Reslan, H., Carano, Invest. 101, 344352. ligand/MMP-9 signaling in prostate
Hruban, R. H., Takaori, K., Klimstra, D. R. A., Kasman, I., Mai, E., Young, J., Kotake, Y., Cao, R., Viatour, P., Sage, cancer. Cancer Res. 66, 48164825.
S., Adsay, N. V., Albores-Saavedra, J., Zha, J., Zhang, Z., Ross, S., Schwall, J., Zhang, Y., and Xiong, Y. (2007). Lian, J. P., Word, B., Taylor, S., Ham-
Biankin, A. V., Biankin, S. A., Comp- R., Colbern, G., and Merchant, M. pRB family proteins are required mons, G. J., and Lyn-Cook, B. D.
ton, C., Fukushima, N., Furukawa, (2008). MetMAb, the one-armed for H3K27 trimethylation and Poly- (2004). Modulation of the consti-
T., Goggins, M., Kato, Y., Klppel, G., 5D5 anti-c-Met antibody, inhibits comb repression complexes binding tutive activated STAT3 transcrip-
Longnecker, D. S., Lttges, J., Maitra, orthotopic pancreatic tumor growth to and silencing p16INK4 tumor tion factor in pancreatic cancer pre-
A., Offerhaus, G. J., Shimizu, M., and and improves survival. Cancer Res. suppressor gene. Genes Dev. 21, vention: effects of indole-3-carbinol
Yonezawa, S. (2004). An illustrated 68, 43604368. 4954. (I3C) and genistein. Anticancer Res.
consensus on the classication of Jones, S., Zhang, X., Parsons, D. W., Lee, C. J., Dosch, J., and Simeone, D. M. 24, 133137.
pancreatic intraepithelial neoplasia Lin, J. C., Leary, R. J., Angenendt, P., (2008). Pancreatic cancer stem cells. Liau, S. S., Ashley, S. W., and Whang, E.
and intraductal papillary mucinous Mankoo, P., Carter, H., Kamiyama, J. Clin. Oncol. 26, 28062812. E. (2006). Lentivirus-mediated RNA
neoplasms. Am. J. Surg. Pathol. 28, H., Jimeno, A., Hong, S. M., Fu, Lee, E. J., Gusev, Y., Jiang, J., Nuovo, interference of HMGA1 promotes
977987. B., Lin, M. T., Calhoun, E. S., G. J., Lerner, M. R., Frankel, W. chemosensitivity to gemcitabine in
Hu, W. G., Liu, T., Xiong, J. X., Kamiyama, M., Walter, K., Nikol- L., Morgan, D. L., Postier, R. G., pancreatic adenocarcinoma. J. Gas-
and Wang, C. Y. (2007). Blockade skaya, T., Nikolsky, Y., Hartigan, J., Brackett, D. J., and Schmittgen, T. trointest. Surg. 10, 12541262; dis-
of sonic hedgehog signal pathway Smith, D. R., Hidalgo, M., Leach, D. (2007). Expression proling iden- cussion 1263.
enhances antiproliferative effect of S. D., Klein, A. P., Jaffee, E. M., ties microRNA signature in pan- Liau, S. S., and Whang, E. (2008).
EGFR inhibitor in pancreatic can- Goggins, M., Maitra, A., Iacobuzio- creatic cancer. Int. J. Cancer 120, HMGA1 is a molecular determinant
cer cells. Acta Pharmacol. Sin. 28, Donahue, C., Eshleman, J. R., Kern, 10461054. of chemoresistance to gemcitabine
12241230. S. E., Hruban, R. H., Karchin, R., Levy, L., and Hill, C. S. (2005). in pancreatic adenocarcinoma. Clin.
Hussain, S. P., Hofseth, L. J., and Har- Papadopoulos, N., Parmigiani, G., Smad4 dependency denes two Cancer Res. 14, 14701477.
ris, C. C. (2003). Radical causes Vogelstein, B., Velculescu, V. E., and classes of transforming growth Liptay, S., Weber, C. K., Ludwig, L., Wag-
of cancer. Nat. Rev. Cancer 3, Kinzler, K. W. (2008). Core signal- factor (TGF-) target genes ner, M., Adler, G., and Schmid, R. M.
276285. ing pathways in human pancreatic and distinguishes TGF--induced (2003). Mitogenic and antiapoptotic
Ischenko, I., Guba, M., Yezhelyev, M., cancers revealed by global genomic epithelial-mesenchymal transition role of constitutive NF-B/Rel activ-
Papyan, A., Schmid, G., Green, T., analyses. Science 321, 18011806. from its antiproliferative and migra- ity in pancreatic cancer. Int. J. Cancer
Fennell, M., Jauch, K. W., and Bruns, Karin, M. (2006). Nuclear factor-B in tory responses. Mol. Cell. Biol. 25, 105, 735746.
C. J. (2007). Effect of Src kinase cancer development and progres- 81088125. Liu, W., Bloom, D. A., Cance, W. G.,
inhibition on metastasis and tumor sion. Nature 441, 431436. Li, L., Aggarwal, B. B., Shishodia, Kurenova, E. V., Golubovskaya, V.
angiogenesis in human pancreatic Kawasaki, D., Emori, Y., Eta, R., Iino, Y., S., Abbruzzese, J., and Kurzrock, M., and Hochwald, S. N. (2008). FAK
cancer. Angiogenesis 10, 167182. Hamano, H., Yoshinaga, K., Tanaka, R. (2004a). Nuclear factor-B and and IGF-IR interact to provide sur-
Itzkowitz, S. H., Yuan, M., Ferrell, L. D., T., Takei, M., and Watson, S. A. IB kinase are constitutively active vival signals in human pancreatic
Ratcliffe, R. M., Chung, Y. S., Satake, (2008). Effect of Z-360, a novel in human pancreatic cells, and adenocarcinoma cells. Carcinogene-
K., Umeyama, K., Jones, R. T., and orally active CCK-2/gastrin recep- their down-regulation by curcumin sis 29, 10961107.
Kim, Y. S. (1987). Cancer-associated tor antagonist on tumor growth (diferuloylmethane) is associated Liu, T. J., LaFortune, T., Honda, T.,
alterations of blood group antigen in human pancreatic adenocarci- with the suppression of prolifera- Ohmori, O., Hatakeyama, S., Meyer,
expression in the human pancreas. J. noma cell lines in vivo and mode tion and the induction of apoptosis. T., Jackson, D., de Groot, J., and Yung,
Natl. Cancer Inst. 79, 425434. of action determinations in vitro. Cancer 101, 23512362. W. K. (2007). Inhibition of both

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 20
Iovanna et al. Pancreatic cancer

focal adhesion kinase and insulin- relation to human disease. N. Engl. Colwell, B., Zee, B., Tu, D., Ott- Nakamura, Y., Migita, T., Hosoda, F.,
like growth factor-I receptor kinase J. Med. 280, 9941006. away, J., Humphrey, R., Seymour, Okada, N., Gotoh, M., Arai, Y.,
suppresses glioma proliferation in Martin, N. E., Brunner, T. B., Kiel, K. D., L., and National Cancer Institute Fukushima, M., Ohki, M., Miyata,
vitro and in vivo. Mol. Cancer Ther. DeLaney, T. F., Regine, W. F., Mohi- of Canada Clinical Trials Group. S., Takeuchi, K., Imoto, I., Katai,
6, 13571367. uddin, M., Rosato, E. F., Haller, D. (2003). Comparison of gemcitabine H., Yamaguchi, T., Inazawa, J., Hiro-
Loakes, D. (2001). Survey and sum- G., Stevenson, J. P., Smith, D., Pra- versus the matrix metalloproteinase hashi, S., Ishikawa, Y., and Shi-
mary: the applications of universal manik, B., Tepper, J., Tanaka, W. K., inhibitor BAY 12-9566 in patients bata, T. (2009). Kruppel-like fac-
DNA base analogues. Nucleic Acids Morrison, B., Deutsch, P., Gupta, A. with advanced or metastatic ade- tor 12 plays a signicant role in
Res. 29, 24372447. K., Muschel, R. J., McKenna, W. G., nocarcinoma of the pancreas: a poorly differentiated gastric can-
Lomberk, G., Wallrath, L., and Urru- Bernhard, E. J., and Hahn, S. M. phase III trial of the National Can- cer progression. Int. J. Cancer 125,
tia, R. (2006). The heterochromatin (2004). A phase I trial of the dual far- cer Institute of Canada Clinical 18591867.
protein 1 family. Genome Biol. 7, 228. nesyltransferase and geranylgeranyl- Trials Group. J. Clin. Oncol. 21, Nakashima, H., Nakamura, M.,
Longnecker, D. S., Adsay, N. V., transferase inhibitor L-778,123 and 32963302. Yamaguchi, H., Yamanaka, N.,
Fernandez-del Castillo, C., Hruban, radiotherapy for locally advanced Mori-Iwamoto, S., Kuramitsu, Y., Akiyoshi, T., Koga, K., Yamaguchi,
R. H., Kasugai, T., Klimstra, D. pancreatic cancer. Clin. Cancer Res. Ryozawa, S., Mikuria, K., Fujimoto, K., Tsuneyoshi, M., Tanaka, M.,
S., Kloppel, G., Luttges, J., Mem- 10, 54475454. M., Maehara, S., Maehara, Y., Okita, and Katano, M. (2006). Nuclear
oli, V. A., Tosteson, T. D., Yanagi- McClintock, B. (1941). The stability of K., Nakamura, K., and Sakaida, I. factor-B contributes to hedge-
sawa, A., Wilentz, R., and Zamboni, broken ends of chromosomes in Zea (2007). Proteomics nding heat hog signaling pathway activation
G. (2005). Histopathological diag- mays. Genetics 26, 234282. shock protein 27 as a biomarker for through sonic hedgehog induction
nosis of pancreatic intraepithelial Milella, M., Gelibter, A., Di Cosimo, resistance of pancreatic cancer cells in pancreatic cancer. Cancer Res. 66,
neoplasia and intraductal papillary- S., Bria, E., Ruggeri, E. M., Car- to gemcitabine. Int. J. Oncol. 31, 70417049.
mucinous neoplasms: interobserver lini, P., Malaguti, P., Pellicciotta, M., 13451350. Nawroth, R., van Zante, A., Cervantes,
agreement. Pancreas 31, 344349. Terzoli, E., and Cognetti, F. (2004). Mori-Iwamoto, S., Taba, K., Kuramitsu, S., McManus, M., Hebrok, M., and
Louvet, C., Labianca, R., Hammel, P., Pilot study of celecoxib and infu- Y., Ryozawa, S., Tanaka, T., Maehara, Rosen, S. D. (2007). Extracellu-
Lledo, G., Zampino, M. G., Andre, sional 5-uorouracil as second-line S., Maehara, Y., Okita, K., Naka- lar sulfatases, elements of the Wnt
T., Zaniboni, A., Ducreux, M., Aitini, treatment for advanced pancreatic mura, K., and Sakaida, I. (2009). signaling pathway, positively regu-
E., Taieb, J., Faroux, R., Lepere, C., carcinoma. Cancer 101, 133138. Interferon- down-regulates heat late growth and tumorigenicity of
de Gramont, A., GERCOR, and GIS- Mimeault, M., Brand, R. E., Sasson, A. shock protein 27 of pancreatic can- human pancreatic cancer cells. PLoS
CAD. (2005). Gemcitabine in com- A., and Batra, S. K. (2005). Recent cer cells and helps in the cytotoxic ONE 2, e392. doi:10.1371/jour-
bination with oxaliplatin compared advances on the molecular mecha- effect of gemcitabine. Pancreas 38, nal.pone.0000392
with gemcitabine alone in locally nisms involved in pancreatic cancer 224226. Neoptolemos, J. P., Stocken, D. D., Tudur
advanced or metastatic pancreatic progression and therapies. Pancreas Morton, J. P., Karim, S. A., Graham, K., Smith, C., Bassi, C., Ghaneh, P.,
cancer: results of a GERCOR and 31, 301316. Timpson, P., Jamieson, N., Athineos, Owen, E., Moore, M., Padbury, R.,
GISCAD phase III trial. J. Clin. Miyamoto, Y., Maitra, A., Ghosh, B., D., Doyle, B., McKay, C., Heung, Doi, R., Smith, D., and Buchler, M.
Oncol. 23, 35093516. Zechner, U., Argani, P., Iacobuzio- M. Y., Oien, K. A., Frame, M. C., W. (2009). Adjuvant 5-uorouracil
Low, S. K., Kuchiba, A., Zembutsu, H., Donahue, C. A., Sriuranpong, V., Evans, T. R., Sansom, O. J., and Brun- and folinic acid vs observation for
Saito, A., Takahashi, A., Kubo, M., Iso, T., Meszoely, I. M., Wolfe, M. ton, V. G. (2010). Dasatinib inhibits pancreatic cancer: composite data
Daigo, Y., Kamatani, N., Chiku, S., S., Hruban, R. H., Ball, D. W., the development of metastases in from the ESPAC-1 and -3(v1) trials.
Totsuka, H., Ohnami, S., Hirose, H., Schmid, R. M., and Leach, S. D. a mouse model of pancreatic duc- Br. J. Cancer 100, 246250.
Shimada, K., Okusaka, T., Yoshida, (2003). Notch mediates TGF - tal adenocarcinoma. Gastroenterol- Nkondjock, A., Ghadirian, P., John-
T., Nakamura, Y., and Sakamoto, H. induced changes in epithelial differ- ogy 139, 292303. son, K. C., and Krewski, D. (2005).
(2010). Genome-wide association entiation during pancreatic tumori- Muerkoster, S., Arlt, A., Witt, M., Gehrz, Dietary intake of lycopene is associ-
study of pancreatic cancer in Japan- genesis. Cancer Cell 3, 565576. A., Haye, S., March, C., Grohmann, ated with reduced pancreatic cancer
ese population. PLoS ONE 5, e11824. Mohammad, R. M., Banerjee, S., Li, F., Wegehenkel, K., Kalthoff, H., risk. J. Nutr. 135, 592597.
doi:10.1371/journal.pone.0011824 Y., Aboukameel, A., Kucuk, O., and Folsch, U. R., and Schafer, H. (2003). Oettle, H., Post, S., Neuhaus, P.,
Lowenfels, A. B., Maisonneuve, P., Cav- Sarkar, F. H. (2006). Cisplatin- Usage of the NF-B inhibitor sul- Gellert, K., Langrehr, J., Ridwel-
allini, G., Ammann, R. W., Lankisch, induced antitumor activity is poten- fasalazine as sensitizing agent in ski, K., Schramm, H., Fahlke, J.,
P. G., Andersen, J. R., Dimagno, E. P., tiated by the soy isoavone genis- combined chemotherapy of pan- Zuelke, C., Burkart, C., Gutberlet,
Andren-Sandberg,A., and Domellof, tein in BxPC-3 pancreatic tumor creatic cancer. Int. J. Cancer 104, K., Kettner, E., Schmalenberg, H.,
L. (1993). Pancreatitis and the risk xenografts. Cancer 106, 12601268. 469476. Weigang-Koehler, K., Bechstein, W.
of pancreatic cancer. International Moore, M. J., Goldstein, D., Hamm, Mukhtar, H., and Ahmad, N. (1999). O., Niedergethmann, M., Schmidt-
Pancreatitis Study Group. N. Engl. J. J., Figer, A., Hecht, J. R., Gallinger, Green tea in chemoprevention of Wolf, I., Roll, L., Doerken, B.,
Med. 328, 14331437. S., Au, H. J., Murawa, P., Walde, cancer. Toxicol. Sci. 52, 111117. and Riess, H. (2007). Adjuvant
Lu, J., Getz, G., Miska, E. A., Alvarez- D., Wolff, R. A., Campos, D., Mund, C., Brueckner, B., and chemotherapy with gemcitabine vs
Saavedra, E., Lamb, J., Peck, D., Lim, R., Ding, K., Clark, G., Lyko, F. (2006). Reactivation observation in patients undergo-
Sweet-Cordero, A., Ebert, B. L., Mak, Voskoglou-Nomikos, T., Ptasynski, of epigenetically silenced genes ing curative-intent resection of
R. H., Ferrando, A. A., Downing, M., Parulekar, W., and National by DNA methyltransferase pancreatic cancer: a randomized
J. R., Jacks, T., Horvitz, H. R., and Cancer Institute of Canada Clini- inhibitors: basic concepts and controlled trial. JAMA 297, 267277.
Golub, T. R. (2005). MicroRNA cal Trials Group. (2007). Erlotinib clinical applications. Epigenetics 1, Ogura, Y., Mizumoto, K., Nagai,
expression proles classify human plus gemcitabine compared with 713. E., Murakami, M., Inadome, N.,
cancers. Nature 435, 834838. gemcitabine alone in patients with Munshi, N., Jeay, S., Li, Y., Chen, C. R., Saimura, M., Matsumoto, K., Naka-
Manolio, T. A. (2010). Genomewide advanced pancreatic cancer: a phase France, D. S., Ashwell, M. A., Hill, J., mura, T., Maemondo, M., Nukiwa,
association studies and assessment III trial of the National Cancer Insti- Moussa, M. M., Leggett, D. S., and T., and Tanaka, M. (2006). Peritu-
of the risk of disease. N. Engl. J. Med. tute of Canada Clinical Trials Group. Li, C. J. (2010). ARQ 197, a novel moral injection of adenovirus vec-
363, 166176. J. Clin. Oncol. 25, 19601966. and selective inhibitor of the human tor expressing NK4 combined with
Marcus, D. M. (1969). The ABO Moore, M. J., Hamm, J., Dancey, c-Met receptor tyrosine kinase with gemcitabine treatment suppresses
and Lewis blood-group system. J., Eisenberg, P. D., Dagenais, M., antitumor activity. Mol. Cancer Ther. growth and metastasis of human
Immunochemistry, genetics and Fields, A., Hagan, K., Greenberg, B., 9, 15441553. pancreatic cancer cells implanted

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 21


Iovanna et al. Pancreatic cancer

orthotopically in nude mice and pro- Zheng, W., Albanes, D., Bamlet, W. PLoS Med. 3, e516. doi:10.1371/jour- Calin, G. A., Volinia, S., Liu, C. G.,
longs survival. Cancer Gene Ther. 13, R., Berg, C. D., Boutron-Ruault, M. nal.pmed.0030516 Scarpa, A., and Croce, C. M. (2006).
520529. C., Buring, J. E., Bracci, P. M., Canz- Poplin, E., Feng, Y., Berlin, J., Rothen- MicroRNA expression abnormali-
Okami, J., Yamamoto, H., Fujiwara, Y., ian, F., Clipp, S., Cotterchio, M., de berg, M. L., Hochster, H., Mitchell, ties in pancreatic endocrine and aci-
Tsujie, M., Kondo, M., Noura, S., Andrade, M., Duell, E. J., Gaziano, E., Alberts, S., ODwyer, P., Haller, nar tumors are associated with dis-
Oshima, S., Nagano, H., Dono, K., J. M., Giovannucci, E. L., Goggins, D., Catalano, P., Cella, D., and tinctive pathologic features and clin-
Umeshita, K., Ishikawa, O., Sakon, M., Hallmans, G., Hankinson, S. E., Benson, A. B. III. (2009). Phase ical behavior. J. Clin. Oncol. 24,
M., Matsuura, N., Nakamori, S., and Hassan, M., Howard, B., Hunter, D. III, randomized study of gemc- 46774684.
Monden, M. (1999). Overexpression J., Hutchinson, A., Jenab, M., Kaaks, itabine and oxaliplatin versus gem- Romero, D., Iglesias, M., Vary, C. P.,
of cyclooxygenase-2 in carcinoma of R., Kooperberg, C., Krogh, V., Kurtz, citabine (xed-dose rate infusion) and Quintanilla, M. (2008). Func-
the pancreas. Clin. Cancer Res. 5, R. C., Lynch, S. M., McWilliams, R. compared with gemcitabine (30- tional blockade of Smad4 leads to a
20182024. R., Mendelsohn, J. B., Michaud, D. S., minute infusion) in patients with decrease in -catenin levels and sig-
Olson, P., Lu, J., Zhang, H., Shai, A., Parikh, H., Patel, A. V., Peeters, P. H., pancreatic carcinoma E6201: a trial naling activity in human pancreatic
Chun, M. G., Wang, Y., Libutti, S. Rajkovic, A., Riboli, E., Rodriguez, of the Eastern Cooperative Oncol- carcinoma cells. Carcinogenesis 29,
K., Nakakura, E. K., Golub, T. R., L., Seminara, D., Shu, X. O., Thomas, ogy Group. J. Clin. Oncol. 27, 10701076.
and Hanahan, D. (2009). MicroRNA G., Tjnneland, A., Tobias, G. S., Tri- 37783785. Ruggeri, B. A., Huang, L., Wood, M.,
dynamics in the stages of tumorige- chopoulos, D., Van Den Eeden, S. Qanungo, S., Das, M., Haldar, S., and Cheng, J. Q., and Testa, J. R. (1998).
nesis correlate with hallmark capa- K., Virtamo, J., Wactawski-Wende, Basu, A. (2005). Epigallocatechin-3- Amplication and overexpression of
bilities of cancer. Genes Dev. 23, J., Wang, Z., Wolpin, B. M., Yu, gallate induces mitochondrial mem- the AKT2 oncogene in a subset
21522165. H., Yu, K., Zeleniuch-Jacquotte, A., brane depolarization and caspase- of human pancreatic ductal ade-
Ouassi, M., Sielezneff, I., Silvestre, R., Fraumeni, J. F. Jr., Hoover, R. N., dependent apoptosis in pancre- nocarcinomas. Mol. Carcinog. 21,
Sastre, B., Bernard, J. P., Lafontaine, Hartge, P., and Chanock, S. J. (2010). atic cancer cells. Carcinogenesis 26, 8186.
J. S., Payan, M. J., Dahan, L., Pirr, A genome-wide association study 958967. Sato, N., Fukushima, N., Maitra,
N., Seitz, J. F., Mas, E., Lom- identies pancreatic cancer suscepti- Ricart, A. D., Tolcher, A. W., Liu, G., A., Matsubayashi, H., Yeo, C. J.,
bardo, D., and Ouaissi, A. (2008). bility loci on chromosomes 13q22.1, Holen, K., Schwartz, G., Albertini, Cameron, J. L., Hruban, R. H., and
High histone deacetylase 7 (HDAC7) 1q32.1 and 5p15.33. Nat. Genet. 42, M., Weiss, G., Yazji, S., Ng, C., and Goggins, M. (2003). Discovery of
expression is signicantly associ- 224228. Wilding, G. (2008). Volociximab, a novel targets for aberrant methyla-
ated with adenocarcinomas of the Petersen, G. M., de Andrade, M., Gog- chimeric monoclonal antibody that tion in pancreatic carcinoma using
pancreas. Ann. Surg. Oncol. 15, gins, M., Hruban, R. H., Bondy, specically binds 51 integrin: a high-throughput microarrays. Can-
23182328. M., Korczak, J. F., Gallinger, S., phase I, pharmacokinetic, and bio- cer Res. 63, 37353742.
Ougolkov, A. V., Bilim, V. N., and Bil- Lynch, H. T., Syngal, S., Rabe, K. logical correlative study. Clin. Cancer Schlieman, M. G., Fahy, B. N., Ram-
ladeau, D. D. (2008). Regulation of G., Seminara, D., and Klein, A. P. Res. 14, 79247929. samooj, R., Beckett, L., and Bold, R.
pancreatic tumor cell proliferation (2006). Pancreatic cancer genetic Rinehart, J., Adjei, A. A., Lorusso, P. M., J. (2003). Incidence, mechanism and
and chemoresistance by the histone epidemiology consortium. Cancer Waterhouse, D., Hecht, J. R., Natale, prognostic value of activated AKT
methyltransferase enhancer of zeste Epidemiol. Biomarkers Prev. 15, R. B., Hamid, O., Varterasian, M., in pancreas cancer. Br. J. Cancer 89,
homologue 2. Clin. Cancer Res. 14, 704710. Asbury, P., Kaldjian, E. P., Gulyas, S., 21102115.
67906796. Philip, P. A., Benedetti, J., Corless, C. Mitchell, D. Y., Herrera, R., Sebolt- Schmitz-Winnenthal, F. H., ZGraggen,
Palmer, D. H., Stocken, D. D., Hewitt, H., L., Wong, R., OReilly, E. M., Flynn, Leopold, J. S., and Meyer, M. B. K., Volk, C., Schmied, B. M., and
Markham, C. E., Hassan, A. B., John- P. J., Rowland, K. M., Atkins, J. N., (2004). Multicenter phase II study of Buchler, M. W. (2003). Intraductal
son, P. J., Buckels, J. A., and Bramhall, Mirtsching, B. C., Rivkin, S. E., Kho- the oral MEK inhibitor, CI-1040, in papillary mucinous tumors of the
S. R. (2007). A randomized phase 2 rana, A. A., Goldman, B., Fenoglio- patients with advanced non-small- pancreas. Curr. Gastroenterol. Rep. 5,
trial of neoadjuvant chemotherapy Preiser, C. M., Abbruzzese, J. L., cell lung, breast, colon, and pan- 133140.
in resectable pancreatic cancer: gem- and Blanke, C. D. (2010). Phase creatic cancer. J. Clin. Oncol. 22, Schutte, M., Hruban, R. H., Geradts, J.,
citabine alone versus gemcitabine III study comparing gemcitabine 44564462. Maynard, R., Hilgers, W., Rabindran,
combined with cisplatin. Ann. Surg. plus cetuximab versus gemcitabine Ringrose, L. (2007). Polycomb comes of S. K., Moskaluk, C. A., Hahn, S. A.,
Oncol. 14, 20882096. in patients with advanced pan- age: genome-wide proling of tar- Schwarte-Waldhoff, I., Schmiegel,
Pan, L. N., Lu, J., and Huang, B. (2007). creatic adenocarcinoma: Southwest get sites. Curr. Opin. Cell Biol. 19, W., Baylin, S. B., Kern, S. E., and
HDAC inhibitors: a potential new Oncology Group-directed inter- 290297. Herman, J. G. (1997). Abrogation
category of anti-tumor agents. Cell. group trial S0205. J. Clin. Oncol. 28, Rocchi, P., Beraldi, E., Ettinger, S., of the Rb/p16 tumor-suppressive
Mol. Immunol. 4, 337343. 36053610. Fazli, L., Vessella, R. L., Nelson, C., pathway in virtually all pancre-
Pasca di Magliano, M., Biankin, A. V., Piao, W., Wang, Y., Adachi, Y., and Gleave, M. (2005). Increased atic carcinomas. Cancer Res. 57,
Heiser, P. W., Cano, D. A., Gutierrez, Yamamoto, H., Li, R., Imsum- Hsp27 after androgen ablation facil- 31263130.
P. J., Deramaudt, T., Segara, D., Daw- ran, A., Li, H., Maehata, T., Ii, M., itates androgen-independent pro- Schwartz, A. M., and Henson, D. E.
son, A. C., Kench, J. G., Henshall, S. Arimura, Y., Lee, C. T., Shinomura, gression in prostate cancer via sig- (2007). Familial and sporadic pan-
M., Sutherland, R. L., Dlugosz, A., Y., Carbone, D. P., and Imai, K. nal transducers and activators of creatic carcinoma, epidemiologic
Rustgi,A. K., and Hebrok, M. (2007). (2008). Insulin-like growth factor-I transcription 3-mediated suppres- concordance. Am. J. Surg. Pathol. 31,
Common activation of canonical receptor blockade by a specic sion of apoptosis. Cancer Res. 65, 645646.
Wnt signaling in pancreatic ade- tyrosine kinase inhibitor for human 1108311093. Seo, Y., Baba, H., Fukuda, T., Takashima,
nocarcinoma. PLoS ONE 2, e1155. gastrointestinal carcinomas. Mol. Rocchi, P., So, A., Kojima, S., Signaevsky, M., and Sugimachi, K. (2000). High
doi:10.1371/journal.pone.0001155 Cancer Ther. 7, 14831493. M., Beraldi, E., Fazli, L., Hurtado- expression of vascular endothelial
Petersen, G. M., Amundadottir, L., Pogue-Geile, K. L., Chen, R., Bron- Coll, A., Yamanaka, K., and Gleave, growth factor is associated with
Fuchs, C. S., Kraft, P., Stolzenberg- ner, M. P., Crnogorac-Jurcevic, T., M. (2004). Heat shock protein 27 liver metastasis and a poor prog-
Solomon, R. Z., Jacobs, K. B., Arslan, Moyes, K. W., Dowen, S., Otey, increases after androgen ablation nosis for patients with ductal pan-
A. A., Bueno-de-Mesquita, H. B., C. A., Crispin, D. A., George, R. and plays a cytoprotective role in creatic adenocarcinoma. Cancer 88,
Gallinger, S., Gross, M., Helzlsouer, D., Whitcomb, D. C., and Brent- hormone-refractory prostate cancer. 22392245.
K., Holly, E. A., Jacobs, E. J., Klein, nall, T. A. (2006). Palladin mutation Cancer Res. 64, 65956602. Shafaee, Z., Schmidt, H., Du, W., Posner,
A. P., LaCroix, A., Li, D., Mandel- causes familial pancreatic cancer and Roldo, C., Missiaglia, E., Hagan, J. P., M., and Weichselbaum, R. (2006).
son, M. T., Olson, S. H., Risch, H. A., suggests a new cancer mechanism. Falconi, M., Capelli, P., Bersani, S., Cyclopamine increases the cytotoxic

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 22
Iovanna et al. Pancreatic cancer

effects of paclitaxel and radiation Takayama, Y., Kokuryo, T., Yokoyama, tumor cells. Clin. Cancer Res. 12, Klein, A. P. (2007). PancPRO: risk
but not cisplatin and gemcitabine Y., Nagino, M., Nimura, Y., Senga, 65576564. assessment for individuals with a
in Hedgehog expressing pancreatic T., and Hamaguchi, M. (2008). MEK Van Cutsem, E., van de Velde, H., family history of pancreatic cancer.
cancer cells. Cancer Chemother. inhibitor enhances the inhibitory Karasek, P., Oettle, H., Vervenne, J. Clin. Oncol. 25, 14171422.
Pharmacol. 58, 765770. effect of imatinib on pancreatic can- W. L., Szawlowski, A., Schoffski, P., Wang, Z., Banerjee, S., Li, Y., Rah-
Shen, Y. M., Yang, X. C., Yang, C., cer cell growth. Cancer Lett. 264, Post, S., Verslype, C., Neumann, man, K. M., Zhang, Y., and Sarkar,
and Shen, J. K. (2008). Enhanced 241249. H., Safran, H., Humblet, Y., Perez F. H. (2006a). Down-regulation of
therapeutic effects for human pan- Talar-Wojnarowska, R., and Malecka- Ruixo, J., Ma, Y., and Von Hoff, D. notch-1 inhibits invasion by inacti-
creatic cancer by application K-ras Panas, E. (2006). Molecular (2004). Phase III trial of gemcitabine vation of nuclear factor-B, vascu-
and IGF-IR antisense oligodeoxynu- pathogenesis of pancreatic ade- plus tipifarnib compared with gem- lar endothelial growth factor, and
cleotides. World J. Gastroenterol. 14, nocarcinoma: potential clinical citabine plus placebo in advanced matrix metalloproteinase-9 in pan-
51765185. implications. Med. Sci. Monit. 12, pancreatic cancer. J. Clin. Oncol. 22, creatic cancer cells. Cancer Res. 66,
Singh, E. K., Ravula, S., Pan, C. M., RA186RA193. 14301438. 27782784.
Pan, P. S., Vasko, R. C., Lapera, S. A., Thayer, S. P., di Magliano, M. P., Van Cutsem, E., Vervenne, W. L., Ben- Wang, Z., Sengupta, R., Banerjee, S.,
Weerasinghe, S. V., Pum, M. K., and Heiser, P. W., Nielsen, C. M., Roberts, nouna, J., Humblet, Y., Gill, S., Van Li, Y., Zhang, Y., Rahman, K. M.,
McAlpine, S. R. (2008). Synthesis D. J., Lauwers, G. Y., Qi, Y. P., Laethem, J. L., Verslype, C., Schei- Aboukameel, A., Mohammad, R.,
and biological evaluation of histone Gysin, S., Fernandez-del Castillo, C., thauer, W., Shang, A., Cosaert, J., Majumdar, A. P., Abbruzzese, J. L.,
deacetylase inhibitors that are based Yajnik, V., Antoniu, B., McMahon, and Moore, M. J. (2009). Phase III and Sarkar, F. H. (2006b). Epidermal
on FR235222: a cyclic tetrapeptide M., Warshaw, A. L., and Hebrok, trial of bevacizumab in combina- growth factor receptor-related pro-
scaffold. Bioorg. Med. Chem. Lett. 18, M. (2003). Hedgehog is an early tion with gemcitabine and erlotinib tein inhibits cell growth and invasion
25492554. and late mediator of pancreatic in patients with metastatic pan- in pancreatic cancer. Cancer Res. 66,
Singh, M., and Maitra, A. (2007). Pre- cancer tumorigenesis. Nature 425, creatic cancer. J. Clin. Oncol. 27, 76537660.
cursor lesions of pancreatic can- 851856. 22312237. Wang, Z., Zhang, Y., Banerjee, S., Li, Y.,
cer: molecular pathology and clin- Tomioka, D., Maehara, N., Kuba, K., Vernejoul, F., Ghenassia, L., Souque, and Sarkar, F. H. (2006c). Inhibition
ical implications. Pancreatology 7, Mizumoto, K., Tanaka, M., Mat- A., Lulka, H., Drocourt, D., Corde- of nuclear factor B activity by genis-
919. sumoto, K., and Nakamura, T. lier, P., Pradayrol, L., Pyronnet, S., tein is mediated via Notch-1 sig-
Sohn, T. A., Yeo, C. J., Cameron, J. L., (2001). Inhibition of growth, inva- Buscail, L., and Tiraby, G. (2006). naling pathway in pancreatic cancer
Iacobuzio-Donahue, C. A., Hruban, sion, and metastasis of human pan- Gene therapy based on gemcitabine cells. Int. J. Cancer 118, 19301936.
R. H., and Lillemoe, K. D. (2001). creatic carcinoma cells by NK4 in chemosensitization suppresses pan- Wang, Z., Zhang, Y., Banerjee, S., Li, Y.,
Intraductal papillary mucinous neo- an orthotopic mouse model. Cancer creatic tumor growth. Mol. Ther. 14, and Sarkar, F. H. (2006d). Notch-
plasms of the pancreas: an increas- Res. 61, 75187524. 758767. 1 down-regulation by curcumin is
ingly recognized clinicopathologic Toubaji, A., Achtar, M., Provenzano, Vire, E., Brenner, C., Deplus, R., Blan- associated with the inhibition of cell
entity. Ann. Surg. 234, 313321; dis- M., Herrin, V. E., Behrens, R., chon, L., Fraga, M., Didelot, C., growth and the induction of apopto-
cussion 321312. Hamilton, M., Bernstein, S., Ven- Morey, L., Van Eynde, A., Bernard, sis in pancreatic cancer cells. Cancer
Sood, P., Krek, A., Zavolan, M., Macino, zon, D., Gause, B., Marincola, F., D., Vanderwinden, J. M., Bollen, M., 106, 25032513.
G., and Rajewsky, N. (2006). Cell- and Khleif, S. N. (2008). Pilot study Esteller, M., Di Croce, L., de Launoit, Wang, Z., Zhang, Y., Li, Y., Banerjee, S.,
type-specic signatures of microR- of mutant ras peptide-based vac- Y., and Fuks, F. (2006). The Poly- Liao, J., and Sarkar, F. H. (2006e).
NAs on target mRNA expression. cine as an adjuvant treatment in comb group protein EZH2 directly Down-regulation of Notch-1 con-
Proc. Natl. Acad. Sci. U.S.A. 103, pancreatic and colorectal cancers. controls DNA methylation. Nature tributes to cell growth inhibition and
27462751. Cancer Immunol. Immunother. 57, 439, 871874. apoptosis in pancreatic cancer cells.
Spano, J. P., Chodkiewicz, C., Maurel, 14131420. Volinia, S., Calin, G. A., Liu, C. G., Mol. Cancer Ther. 5, 483493.
J., Wong, R., Wasan, H., Barone, C., Trapasso, F., Sarti, M., Cesari, R., Yen- Ambs, S., Cimmino, A., Petrocca, F., Wang, Z., Ma, Q., Liu, Q., Yu, H.,
Letourneau, R., Bajetta, E., Pithavala, damuri, S., Dumon, K. R., Aqeilan, Visone, R., Iorio, M., Roldo, C., Fer- Zhao, L., Shen, S., and Yao, J.
Y., Bycott, P., Trask, P., Liau, K., R. I., Pentimalli, F., Infante, L., Alder, racin, M., Prueitt, R. L., Yanaihara, (2008). Blockade of SDF-1/CXCR4
Ricart, A. D., Kim, S., and Rixe, O. H., Abe, N., Watanabe, T., Vigli- N., Lanza, G., Scarpa, A., Vecchione, signalling inhibits pancreatic can-
(2008). Efcacy of gemcitabine plus etto, G., Croce, C. M., and Fusco, A. A., Negrini, M., Harris, C. C., and cer progression in vitro via inactiva-
axitinib compared with gemcitabine (2004). Therapy of human pancre- Croce, C. M. (2006). A microRNA tion of canonical Wnt pathway. Br. J.
alone in patients with advanced pan- atic carcinoma based on suppression expression signature of human solid Cancer 99, 16951703.
creatic cancer: an open-label ran- of HMGA1 protein synthesis in pre- tumors denes cancer gene targets. Watanabe, S., Ueda, Y., Akaboshi, S.,
domised phase II study. Lancet 371, clinical models. Cancer Gene Ther. Proc. Natl. Acad. Sci. U.S.A. 103, Hino, Y., Sekita, Y., and Nakao,
21012108. 11, 633641. 22572261. M. (2009). HMGA2 maintains
Su, G. H., Hruban, R. H., Bansal, R. K., Trevino, J. G., Summy, J. M., Lesslie, D. Wan, J., Xia, Y., Liu, Y., Wang, M., Roc- oncogenic RAS-induced epithelial-
Bova, G. S., Tang, D. J., Shekher, M. P., Parikh, N. U., Hong, D. S., Lee, chi, P., Yao, J., Qu, F., Neyts, J., mesenchymal transition in human
C., Westerman, A. M., Entius, M. M., F. Y., Donato, N. J., Abbruzzese, J. Iovanna, J. L., and Peng, L. (2009). pancreatic cancer cells. Am. J. Pathol.
Goggins, M.,Yeo, C. J., and Kern, S. E. L., Baker, C. H., and Gallick, G. E. Discovery of novel arylethynyltria- 174, 854868.
(1999). Germline and somatic muta- (2006). Inhibition of SRC expression zole ribonucleosides with selective Wei, D., Wang, L., He, Y., Xiong, H.
tions of the STK11/LKB1 Peutz- and activity inhibits tumor progres- and effective antiviral and antipro- Q., Abbruzzese, J. L., and Xie, K.
Jeghers gene in pancreatic and bil- sion and metastasis of human pan- liferative activity. J. Med. Chem. 52, (2004). Celecoxib inhibits vascular
iary cancers. Am. J. Pathol. 154, creatic adenocarcinoma cells in an 11441155. endothelial growth factor expres-
18351840. orthotopic nude mouse model. Am. Wang, W., Abbruzzese, J. L., Evans, D. sion in and reduces angiogenesis and
Szafranska, A. E., Davison, T. S., John, J. Pathol. 168, 962972. B., and Chiao, P. J. (1999). Over- metastasis of human pancreatic can-
J., Cannon, T., Sipos, B., Maghnouj, Tsuda, N., Ishiyama, S., Li, Y., Ioan- expression of urokinase-type plas- cer via suppression of Sp1 transcrip-
A., Labourier, E., and Hahn, S. A. nides, C. G., Abbruzzese, J. L., minogen activator in pancreatic ade- tion factor activity. Cancer Res. 64,
(2007). MicroRNA expression alter- and Chang, D. Z. (2006). Syn- nocarcinoma is regulated by consti- 20302038.
ations are linked to tumorigene- thetic microRNA designed to target tutively activated RelA. Oncogene 18, Wei, Y., Xia, W., Zhang, Z., Liu, J., Wang,
sis and non-neoplastic processes in glioma-associated antigen 1 tran- 45544563. H., Adsay, N. V., Albarracin, C., Yu,
pancreatic ductal adenocarcinoma. scription factor inhibits division and Wang, W., Chen, S., Brune, K. A., D., Abbruzzese, J. L., Mills, G. B.,
Oncogene 26, 44424452. induces late apoptosis in pancreatic Hruban, R. H., Parmigiani, G., and Bast, R. C. Jr., Hortobagyi, G. N.,

www.frontiersin.org January 2012 | Volume 2 | Article 6 | 23


Iovanna et al. Pancreatic cancer

and Hung, M. C. (2008). Loss of an increasingly common diagnosis. pancreatic ductal adenocarcino- growth factor receptor-related
trimethylation at lysine 27 of his- Ann. Surg. 243, 673680; discussion mas with widespread microsatel- protein inhibits cell growth and
tone H3 is a predictor of poor out- 680673. lite instability. Cancer Res. 61, induces apoptosis of BxPC3 pan-
come in breast, ovarian, and pan- Wolpin, B. M., Hezel, A. F., Abrams, 31393144. creatic cancer cells. Cancer Res. 65,
creatic cancers. Mol. Carcinog. 47, T., Blaszkowsky, L. S., Meyerhardt, Zeng, G., Germinaro, M., Micsenyi, A., 38773882.
701706. J. A., Chan, J. A., Enzinger, P. C., Monga, N. K., Bell, A., Sood, A.,
Whitcomb, D. C., Gorry, M. C., Preston, Allen, B., Clark, J. W., Ryan, D. P., Malhotra, V., Sood, N., Midda, V.,
R. A., Furey, W., Sossenheimer, M. and Fuchs, C. S. (2009). Oral mTOR Monga, D. K., Kokkinakis, D. M., Conict of Interest Statement: The
J., Ulrich, C. D., Martin, S. P., Gates, inhibitor everolimus in patients with and Monga, S. P. (2006). Aberrant authors declare that the research was
L. K. Jr., Amann, S. T., Toskes, P. P., gemcitabine-refractory metastatic Wnt/-catenin signaling in pancre- conducted in the absence of any com-
Liddle, R., McGrath, K., Uomo, G., pancreatic cancer. J. Clin. Oncol. 27, atic adenocarcinoma. Neoplasia 8, mercial or nancial relationships that
Post, J. C., and Ehrlich, G. D. (1996). 193198. 279289. could be construed as a potential con-
Hereditary pancreatitis is caused by a Xia, Y., Liu, Y., Wan, J., Wang, M., Zhang, H., Morisaki, T., Nakahara, C., ict of interest.
mutation in the cationic trypsinogen Rocchi, P., Qu, F., Iovanna, J. L., Matsunaga, H., Sato, N., Nagumo,
gene. Nat. Genet. 14, 141145. and Peng, L. (2009). Novel triazole F., Tadano, J., and Katano, M. Received: 19 September 2011; accepted:
Wilentz, R. E., Goggins, M., Redston, M., ribonucleoside down-regulates heat (2003). PSK-mediated NF-B inhi- 11 January 2012; published online: 31
Marcus, V. A., Adsay, N. V., Sohn, T. shock protein 27 and induces potent bition augments docetaxel-induced January 2012.
A., Kadkol, S. S., Yeo, C. J., Choti, anticancer activity on drug-resistant apoptosis in human pancreatic can- Citation: Iovanna J, Mallmann MC,
M., Zahurak, M., Johnson, K, Tas- pancreatic cancer. J. Med. Chem. 52, cer cells NOR-P1. Oncogene 22, Gonalves A, Turrini O and Dagorn J-
cilar, M, Offerhaus, G. J., Hruban, R. 60836096. 20882096. C (2012) Current knowledge on pan-
H., and Kern, S. E. (2000). Genetic, Yachida, S., Jones, S., Bozic, I., Antal, Zhang, Y., Banerjee, S., Wang, Z., creatic cancer. Front. Oncol. 2:6. doi:
immunohistochemical, and clinical T., Leary, R., Fu, B., Kamiyama, Xu, H., Zhang, L., Mohammad, R., 10.3389/fonc.2012.00006
features of medullary carcinoma of M., Hruban, R. H., Eshleman, Aboukameel, A., Adsay, N. V., Che, This article was submitted to Frontiers
the pancreas: a newly described and J. R., Nowak, M. A., Velculescu, M., Abbruzzese, J. L., Majumdar, A. in Molecular and Cellular Oncology, a
characterized entity. Am. J. Pathol. V. E., Kinzler, K. W., Vogelstein, P., and Sarkar, F. H. (2006). Anti- specialty of Frontiers in Oncology.
156, 16411651. B., and Iacobuzio-Donahue, C. A. tumor activity of epidermal growth Copyright 2012 Iovanna, Mallmann,
Winter, J. M., Cameron, J. L., Lille- (2010). Distant metastasis occurs factor receptor-related protein is Gonalves, Turrini and Dagorn. This is
moe, K. D., Campbell, K. A., Chang, late during the genetic evolution mediated by inactivation of ErbB an open-access article distributed under
D., Riall, T. S., Coleman, J., Sauter, of pancreatic cancer. Nature 467, receptors and nuclear factor-B in the terms of the Creative Commons Attri-
P. K., Canto, M., Hruban, R. H., 11141117. pancreatic cancer. Cancer Res. 66, bution Non Commercial License, which
Schulick, R. D., Choti, M. A., and Yamamoto, H., Itoh, F., Nakamura, H., 10251032. permits non-commercial use, distribu-
Yeo, C. J. (2006). Periampullary Fukushima, H., Sasaki, S., Perucho, Zhang, Y., Banerjee, S., Wang, Z. W., tion, and reproduction in other forums,
and pancreatic incidentaloma: a M., and Imai, K. (2001). Genetic Marciniak, D. J., Majumdar, A. P., provided the original authors and source
single institutions experience with and clinical features of human and Sarkar, F. H. (2005). Epidermal are credited.

Frontiers in Oncology | Molecular and Cellular Oncology January 2012 | Volume 2 | Article 6 | 24

Potrebbero piacerti anche