Recent Advances in Pancreatic Cancer: Biology, Treatment, and Prevention Divya Singh, Ghanshyam Upadhyay, Rakesh K. Srivastava, Sharmila Shankar PII: DOI: Reference:

S0304-419X(15)00035-9 doi: 10.1016/j.bbcan.2015.04.003 BBACAN 88039

To appear in:

BBA - Reviews on Cancer

Received date: Revised date: Accepted date:

30 January 2015 28 April 2015 30 April 2015

Please cite this article as: Divya Singh, Ghanshyam Upadhyay, Rakesh K. Srivastava, Sharmila Shankar, Recent Advances in Pancreatic Cancer: Biology, Treatment, and Prevention, BBA - Reviews on Cancer (2015), doi: 10.1016/j.bbcan.2015.04.003

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

PT

Recent Advances in Pancreatic Cancer: Biology, Treatment, and Prevention

SC

RI

Divya Singh1, Ghanshyam Upadhyay1*, Rakesh K. Srivastava2*, Sharmila Shankar2,3*

1

Department of Biology, City College of New York, 160 Convent Avenue, New York, NY-10031 Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128.

NU

2 3

TE

D

MA

Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108.

Email:

AC CE P

[email protected] [email protected]

[email protected] [email protected]

*Corresponding author: Dr. Rakesh Srivastava ([email protected]) Dr. Sharmila Shankar ([email protected])

ACCEPTED MANUSCRIPT Abstract Pancreatic cancer (PC) is the fourth leading cause of cancer-related death in United

PT

States. Efforts have been made towards the development of the viable solution for its

RI

treatment with constrained accomplishment because of its complex biology. It is well

SC

established that pancreatic cancer stem cells (CSCs), albeit present in a little count, contribute incredibly to PC initiation, progression, and metastasis. Customary chemo and

NU

radiotherapeutic alternatives, however, expands general survival, the related side effects

MA

are the significant concern. Amid the most recent decade, our insight about molecular and cellular pathways involved in PC and role of CSCs in its progression has increased

D

enormously. Presently the focus is to target CSCs. The herbal products have got much

TE

consideration recently as they, usually, sensitize CSCs to chemotherapy and target molecular signaling involved in various tumors including PC. Some planned studies have

AC CE P

indicated promising results proposing that examinations in this course have a lot to offer for the treatment of PC. Although preclinical studies uncovered the importance of herbal products in attenuating pancreatic carcinoma, limited studies have been conducted to evaluate their role in clinics. The present review provides a new insight to recent advances in pancreatic cancer biology, treatment and current status of herbal products in its anticipation.

ACCEPTED MANUSCRIPT Abbreviations: Oct4: octamer-binding transcription factor 4; ABCG2: ATP-binding cassette sub-family

PT

G member 2; CXCR4; C-X-C chemokine receptor type 4; FGF: fibroblast growth factor;

RI

Frizzled-9: frizzled class receptor 9; Glut1: glucose transporter 1; Foxa2: forkhead box

SC

A2; Sox2: sex determining region Y box 2; Klf4: kruppel like factor 4; c-Myc: v-Myc avian myelocytomatosis viral oncogene homolog; FGF; fibroblast growth factor; ESA:

NU

epithelial-specific antigen; ALDH1: acetaldehyde dehydrogenases 1; ABCB1: ATP-

MA

binding cassette sub-family B member 1; MDR1: multidrug resistance protein 1; DCLK1: doublecortin-like kinase 1; Cdkn2a: cyclin-dependent kinase inhibitor 2a; Dpc4

D

or Smad4: deleted in pancreatic carcinoma, locus 4; STAT3: signal transducer and

TE

activator of transcription 3; TNF-α: tumor necrosis factor α; MCP-1: monocyte chemotactic protein-1; EGF: epidermal growth factor; EGFR: epidermal growth factor

factor;

AC CE P

receptor; PDGF: platelet-derived growth factor; G-CSF: granulocyte colony-stimulating GM-CSF:

granulocyte-macrophage

colony-stimulating

factor;

TGF-β

:

transforming growth factor beta; Chk2: checkpoint kinase 2; COX-2: cyclooxygenase-2; IGF-1R: insulin-like growth factor-1 receptor; VEGF: vascular endothelial growth factor; HIF1α: hypoxia inducible factor 1α; MMP: matrix metalloproteinase, TWIST1: Twistrelated protein 1; ICAM1: intercellular adhesion molecule 1; Bcl-2: B-cell lymphoma 2, Bcl-xL: B-cell lymphoma extra-large, Bad: Bcl-2-associated death promoter; Bak: Bcl-2 homologous antagonist/killer; Bax: Bcl-2-associated X protein; Mcl-1: induced myeloid leukemia cell differentiation protein; Pdx1: pancreatic and duodenal homeobox 1, uPA: urokinase-type plasminogen activator; uPAR: urokinase-type plasminogen activator receptor; MAPK: mitogen activated protein kinase; FoxO1: forkhead box O1; FoxO3:

ACCEPTED MANUSCRIPT forkhead box O3; PI3K: phosphatidylinositol 3-kinase; PARP: peroxisome proliferatoractivated receptor; PTEN: phosphatase and tensin homolog; PDGFRα: alpha-type

PT

platelet-derived growth factor receptor; IGF2R: insulin-like growth factor 2 receptor;

RI

ENG: endoglin, ALK1: activin receptor-like kinase 1; FKHRL1: forkhead box O3a; FKHR: forkhead box O1: AFX: forkhead box O4; TP53: tumor protein p53; TRAIL:

SC

tumor necrosis factor-related apoptosis-inducing ligand; Cdk4: cyclin-dependent kinase

NU

4; Raf-1: RAF proto-oncogene serine/threonine-protein kinase; Her-2: human epidermal growth factor receptor 2; EMT: endothelial to meseanchymal transition; DR: death

MA

receptor; EpCAM: epithelial cell adhesion molecule; vWF: von Willebrand factor; PCNA: proliferating cell nuclear antigen; Hsp: heat shock potein; XIAP: X-linked apoptosis

protein;

IAP:

D

of

inhibitor

of

apoptosis

protein;

Pdk1:

TE

inhibitor

phosphoinositide-dependent kinase-1; mTOR: mammalian target of rapamycin; ERK:

AC CE P

extracellular-signal-regulated kinases; JNK: c-Jun N-terminal kinases; HDAC: histone deacetylases; p38: P38 mitogen-activated protein kinases; ROS: reactive oxygen species

ACCEPTED MANUSCRIPT Introduction The burden of pancreatic cancer (PC) has continuously increased worldwide. It is a

PT

serious health concern and fourth leading cause of cancer-related death in United States

RI

of America [1, 2]. PC is described as a type of gastrointestinal tumor with a poor

SC

anticipation and a high level of danger and death rate [3]. More than 90% of pancreatic tumors have inception from the ductal epithelium of pancreas consequently termed as

NU

pancreatic ductal adenocarcinoma (PDAC). It is disturbing to see that frequency rate of

MA

the pancreatic tumor is relentlessly expanding in the western world [4]. The danger components for pancreatic growth incorporate smoking, obesity and high utilization of

D

processed meat. Age is positively correlated with pancreatic cancer incidences, and the

TE

larger part of cases are diagnosed over the age of 60 [5]. The introductory indications of patients with PDAC are back agony and dyspepsia with additional disturbing

AC CE P

manifestations like the new onset of diabetes, jaundice, and unconstrained profound vein thrombosis and weight reduction. When one begins perceiving, the tumor typically, spreads to the encompassing tissues or distant organs. For the tumors spotted in the head region of pancreas, the determination is actually productive and they are diagnosed relatively early because of biliary impediment. Nonetheless, the tumors in the body and tail of pancreas regularly stay asymptomatic until late in disease stage. Most of the patients (~80%) are identified with unresectable locally advanced or metastatic disease and the major cause is the delayed diagnosis and lack of specific blood or urine biomarkers to identify patients with increased risk of developing pancreatic cancer [6-9]. The routine diagnostics incorporate transabdominal ultrasound in the introductory

ACCEPTED MANUSCRIPT assessment of the jaundiced patient alongside computed tomography (CT) scan or magnetic resonance imaging (MRI).

PT

Despite the fact that the survival rate for most diseases has been increased lately in a

RI

couple of decades, little change is seen in the case of pancreatic cancer. The usual

SC

survival rate for pancreatic disease patients is under six months, and just 3% patients survive over 5-years [6-9]. The reason is attributed to various factors including silent

NU

nature in early stages, aggressive tumor biology, the low scope of surgical management,

MA

and lack of effective systemic therapies. Although, the current procedures including surgery, chemotherapy, radiation, and immunosuppressants, have made great advances in

D

diminishing tumor frequencies and death rates, pancreatic cancer remains a continuing

TE

challenge to the researchers and treatment strategies at present utilized are not very encouraging [10]. There are exceptionally poor post-surgery survival rates even when the

AC CE P

pancreatic tumor is surgically resected. Safety concerns related with these medications/techniques are likewise a significant issue for their accomplishment in the treatment of the disease [6-9]. The prevalent chemotherapeutic choices for the cancer treatment prolong the life of pancreatic cancer patients minimally, and the survival span in a large portion of the cases is not over one year. Since limited treatment choices are accessible, and it additionally shows resistance against chemo- and radiotherapies, it is important to find novel and viable methodologies for the treatment of pancreatic cancer [10]. Although the potential use of herbal components for the protection against various cancers began several decades ago, studies to understand the mechanism of their action at biochemical, genomic, and proteomic levels started very recently. Many plant products

ACCEPTED MANUSCRIPT that are rich sources of phytochemicals, such as triterpenes, flavonoids or polyphenols, are now established potent chemopreventive agents [11-15]. The phenolic substances are

PT

isolated from the wide range of vascular plants and have the ability to reduce and

RI

scavenge free radicals [16, 17]. Epidemiological studies have shown the reduced risk of pancreatic cancer by increased consumption of fruits and vegetables [18]. In the recent

SC

past, a number of preclinical studies have demonstrated various degrees of the efficacy of

NU

herbal products both in vitro and in vivo [18]. Certain dietary agents, for example, resveratrol and curcumin, have been demonstrated to potentiate the standard

MA

chemotherapy [18]. It has been observed that herbal products target different pathways simultaneously therefore any solution including these products may be a smart thought

TE

D

for better results. Many groups are working in this direction, and the outcomes are promising towards the improvement of new helpful cure. In this review, we will examine

AC CE P

the biology of the pancreatic tumor, diagnosis, treatment techniques and clinical trials. We will likewise concentrate on the plausible role of herbal products, alone or in combination with systemic chemopreventive medications, in the treatment of the pancreatic tumor.

Biology of Pancreatic Cancer The biology of pancreatic cancer is perplexing and inadequately caught on. Pancreas has both exocrine and endocrine cells that can structure tumors; however, the likelihood is more for exocrine cells. The vast majority of the exocrine tumors are adenocarcinomas that begin in organ cells in the ductal epithelium and advances from premalignant injuries to the entirely invasive tumor. Tumors of the endocrine pancreas, commonly termed as islet cell tumors or neuroendocrine tumors, are less common and can be characterized

ACCEPTED MANUSCRIPT into gastrinomas, insulinomas, glucagonomas, somatostatinomas, VIPomas, PPomas and so forth.

PT

The microenvironment of the pancreatic tumor is made out of a few components, for

RI

example, pancreatic cancer bulk cells, pancreatic cancer stem cells (pancreatic CSCs),

SC

and the thick, ineffectively vascularized stroma. The studies suggest that the stroma likewise regulate the pancreatic tumor growth and development, intrusion and metastasis

NU

separated from its movement as the mechanical boundary. These stromal cells

MA

consistently interface with cancer cells by different autocrine and paracrine secretion of the pancreas, for example, platelet-derived growth factor (PDGF), transforming growth

D

factor β (TGF-β) and cytokines [19]. These development variables fortify a critical

TE

segment of stroma called pancreatic stellate cells, which in turn express α–smoothmuscle actin and produce rich collagen fibers. These fibers add to tumor hypoxia (a

AC CE P

primary stimulator of tumor movement and metastasis by influencing angiogenesis), cell survival, and apoptotic pathways [19]. Hypoxia is likewise proposed to be a major cause for drug/therapy resistance in different tumors [20, 21]. Pancreatic tumor cells apparently grow around a population of cancer stem cells (CSCs) that have the capability of self-renewal and multi-lineage differentiation [22] (Fig 1). Pancreatic CSCs can be isolated by flow cytometry utilizing CD44, CD24, and ESA as surface markers [23]. Although CD44+/CD24+/ESA+ cells constitute only 0.2-0.8% of the total cell population, they are capable of forming tumor spheres [24]. Other markers for pancreatic CSCs are CD133, CXCR4, c-Met and ALDH1 [25-28]. CXCR4 assumes an essential part in the tumor invasion and metastasis and the cells positive for both CD133 and CXCR4, show higher metastatic potential than other populations [26, 29]. In

ACCEPTED MANUSCRIPT a recent study, it has been demonstrated that human PDACs contain CSCs with high levels of CXCR4 and ABCB1, and such patients had reduced survival rate [30]. Recently

PT

Shankar et al. effectively demonstrated the tumorigenic potential of pancreatic CSCs

RI

isolated from human pancreatic tumors in NOD/SCID mice utilizing surface markers CD44, ESA, CD133, and CD24 [10]. These cells were highly tumorigenic and were

SC

likewise expressing ALDH and pluripotency maintaining factor, Oct-4 [10]. They further

NU

indicated high expression of CD133, CD24, CD44, ESA, Nanog, Notch1, MDR1 and ABCG2 in these CSCs (CD133+CD44+CD24+ESA+ cell population) contrasted with

MA

CD133−CD44−CD24−ESA− cell population [10].

D

Various elements regulate the properties and conduct of pancreatic CSCs, for example,

TE

nestin can balance attack or metastasis of pancreatic CSCs, Oct-4 and Nanog can direct pancreatic CSC’s conduct and metastasis to different organs, DCLK1 can segregate

AC CE P

between normal and tumoral stem cells and Sox2 controls cell proliferation and differentiation [31]. Furthermore, c-Kit and Kras likewise tweak the movement of pancreatic adenocarcinoma [32]. Epithelial to mesenchymal transition (EMT), a process of change of epithelial attributes into mesenchymal properties, is an urgent process for tumor progression and is proposed to be in charge for the appearance of cells with stem cell-like properties [33]. CSCs are thought to be the major contributory element to the absence of effective treatment for pancreatic malignancy and are in charge of tumorigenesis, metastasis, and development of chemo and radioresistance [24]. A recent study exhibited that PANC-1 cancer cell line, steadily overexpressing Oct4 and Nanog, show chemoresistance, multiplication, relocation, intrusion, and tumorigenesis in vitro and in vivo [34].

ACCEPTED MANUSCRIPT Moreover, the ALDH+CD44+CD24+ cell population is well reported to be impervious to treatment with gemcitabine [35]. Removal of CSCs is essential for robust tumor

RI

be a superior alternative for pancreatic cancer prevention.

PT

treatment, as CSCs stay untouched. Hence, drugs that can specifically target CSCs could

SC

Pancreatic cancer is hereditarily complex and heterogeneous in nature. Different malady conditions, for example, pancreatitis, cystic fibrosis, and inflammation have their effect

NU

on the initiation of pancreatic cancer and its malignant progression [36, 37]. Mutations in

MA

four critical genes namely Kras2, Cdkn2a, TP53 and Dpc4 (or Smad4) are frequently seen in pancreatic malignancy patients. The vast majority of the cancer patients carry one

D

or more of these mutations [38]. The observed frequency of Kras mutation is more than

TE

90%, however, the rate of inactivation mutation of Cdkn2a, TP53, and Dpc4 are 95%, 75% and 50% respectively [39, 40]. The mutation in Kras2 brings about the consistent

AC CE P

expression of irregular Ras protein that causes aberrant activation of cell proliferation and survival pathways [19]. With increasing age, the likelihood of acquiring activating mutations in Kras2 gene increases in the major organs such as lung, pancreas, colon, and other tissues. On the other hand Cdkn2a, TP53 and Dpc4 are the tumor suppressors and mutations in these genes result in their inactivation, which facilitate the proliferation and survival signaling [19, 41]. Moreover, a recent study called attention to the loss of function mutation in SWI/SNF nucleosome remodeling complex in 23% pancreatic adenocarcinomas [42]. Signaling Pathways in Pancreatic Cancer

ACCEPTED MANUSCRIPT Genetic mutations serve as the basis for abbrent signaling pathways. A comprehensive study with more than 24 pancreatic cancer cases, an average of 63 relevent genetic

PT

abnormalities (mainly point mutations) per tumor were classified as likely to be relevent

RI

in its pathogenesis [43] (reviewed in [19]). These mutations can be clubbed in 12 noteworthy signaling pathways including STAT3, Smad/TGF-β, Wnt, Notch, PI3K/Akt,

SC

sonic hedgehog and so forth [43] (reviewed in [19]). Aberrant signaling in these cellular

NU

events has been implicated in the development and progression of pancreatic tumors by

MA

permitting increased proliferation, angiogenesis, survival, and metastasis (Fig 2). STAT3 Pathway

D

Signal transducer and activator of transcription 3 (STAT3), encoded by STAT3 gene, is

TE

activated in a wide variety of signaling pathways. It mediates diverse responses,

AC CE P

including transmission of the signals of cytokines and growth factors from the cell membrane to the nucleus to regulate gene expression for cell development, differentiation, proliferation, survival, and angiogenesis [44-46] (Fig 3). A range of cytokines including interleukin (IL)-6, IL-9, IL-10, IL-27, tumor necrosis factor α (TNFα), and monocyte chemotactic protein-1 (MCP-1) activate the STAT3 pathway [47-49]. Various growth factors, for example, epidermal growth factor (EGF), platelet-derived growth factor (PDGF), granulocyte colony-stimulating factor (G-CSF), and granulocytemacrophage colony-stimulating factor (GM-CSF) also activate this pathway [44-49]. Primary functions of activated STAT3 pathway incorporate cell proliferation by upregulating cyclin D1 and cyclin B1 and inhibition of apoptosis by up-regulating Bcl-2, Bcl-xL, Mcl-1. Initiated STAT3 assumes a discriminating part in tumorigenesis by

ACCEPTED MANUSCRIPT controlling angiogenesis (VEGF, FGF, and HIF1α), invasion and metastasis (MMP2, MMP9, TWIST, and ICAM1) [44-49].

PT

STAT3 activation has been detected in diverse type of malignancies, and its inhibition by

RI

use of inhibitors or short interfering RNA has prompted to reverse the malignant

SC

phenotype. STAT3 activation has been portrayed in almost 70% of solid and hematological malignancies [50]. Studies in conditional knockout mice have

NU

demonstrated that STAT3 pathway is latent in typical pancreas and is not needed for any

MA

vital process related to pancreatic development and homeostasis [51]. Nevertheless, it is constitutively activated in PDAC by phosphorylation of Tyr705 in human tumor

D

specimens and also in various PDAC cell lines [52-55]. Further, STAT3 is necessary for

TE

the development of ADM process (acinar-to-ductal metaplasia), which is an early event in PDAC pathogenesis mediated by ectopic expression of the Pdx1 [54]. It is remarkable

AC CE P

that Pdx1 is a transcription factior and a key regulator of early pancreatic development [54]. Another study showed that with malignant transformation, activated STAT3 promotes proliferation of cells by regulating G1/S-phase progression and supports the malignant phenotype of human pancreatic cancer [52]. IL-6 signaling dependent activation of STAT3 plays an important role in promoting PanIN progression and the PDAC development, in addition to oncogenic KrasG12D transformation [56]. The myeloid cells in the pancreas induce STAT3 activation by releasing IL-6, which promote PanIN progression and the PDAC development. Smad/TGF-β Pathway

ACCEPTED MANUSCRIPT The transforming growth factor beta (TGF-β) signaling pathway regulates various cellular processes like cell growth, cell differentiation, apoptosis and cellular homeostasis

PT

in both the adult organism and the developing embryo [57]. TGF-β signaling occurs from

RI

the membrane to the nucleus via Smad proteins [58]. Smads can be classified into 3 major groups; receptor-regulated Smads (R-Smad; Smad1, Smad2, Smad3, Smad5 and

[58]. Cascade is triggered by the binding of TGF-β

NU

(I-Smad; Smad6 and Smad7)

SC

Smad8/9), common-mediator Smad (co-Smad; Smad4) and agonistic or inhibitory Smads

superfamily ligand to a type II receptor which catalyzes the recruitment and

MA

phosphorylation of a type I receptor. Subsequently R-Smads are phosphorylated, linked with the coSmad and other factors, and finally accumulated in the nucleus to regulate the

TE

D

target gene expression. TGF-β receptor activation results in Smad2 and Smad3 phosphorylation, which then form heteromeric complexes with Smad4. Furthermore,

AC CE P

Smad6 and Smad7, can prevent TGF-β signaling by interacting either with the receptor or with Smad2 and Smad3 [58].

TGF-β signaling pathway impairment because of inactivated Smad4 (DPC4) is often recognized in pancreatic carcinomas [59, 60]. Jonson et al., investigated a series of pancreatic carcinoma cell lines with respect to alterations of five Smad genes involved in TGF-β signaling, and demonstrated the structural rearrangement of Smad4 in 42% of these tumor cells [59]. Since, this pathway could likewise be influenced by other factors that regulate the activation of TGF-β

and its receptor genes, they further assessed

expression of uPA, uPAR, IGF2R, TGF-βR1-3, ENG, ALK1, TGF-β1-3, mutations of TGF-βR1-2, cell surface localization of TGF-βR2 and ENG, and TGF-β1 response in 14 pancreatic carcinoma cell lines [59]. The study suggested ALK5- Smad4 as a major

ACCEPTED MANUSCRIPT target for inactivation in pancreatic carcinomas and that the expression of TGF-βR2, TGF-βR3, and receptors involved in TGF-β activation are maintained [59].

PT

Wnt Pathway

RI

Wnt signaling pathway is a complex process and plays an important role in tumor

SC

development apart from its involvement in other physiological and pathological processes

NU

[61]. In canonical pathway, the ligand binding to its receptor (Frizzled/LRP receptor complexes) triggers a cascade of events that prevents β-catenin degradation inside the

MA

cytoplasm and permits its stabilization and translocation in nucleus where it binds to transcriptional factors of the Tcf/Lef family forming an activator complex. Abnormal

D

Wnt/β-catenin signaling is reported in pancreatic cancer [62]. Activated Wnt signal leads

TE

to the accumulation of β-catenin in the nucleus where it activates specific target genes

AC CE P

[63]. The accumulation of β-catenin is observed both in nucleus and cytoplasm in pancreatic cancer [61, 64-66]. The functional evidences are also accumulating that implicate a supporting role for β-catenin in PDAC maintenance and progression [61]. It has been also found that β-catenin accumulation and signaling could be increased through paracrine signaling taking place in the PDAC micro-environment [61]. In a recent investigation, it has been found that Wnt/β-catenin signaling inhibition by wnt-c59 results in reversal of TSA sensitivity, migration ability, and the EMT phenotype in trichostatin A-resistant Panc-1 cells (Panc-1/TSA) [67]. In spite of the fact that our understanding of the role of this pathway has increased during the last decade, the general mechanism by which β-catenin accumulation occurs in PDAC is poorly understood and needs further elucidation.

ACCEPTED MANUSCRIPT Notch Pathway Notch signaling is shown to regulate proliferation and apoptosis events in various cell

PT

types. The alterations in Notch signaling have various consequences including

RI

tumorigenesis [68, 69]. In mammals, four Notch receptors (Notch1-4) and five ligands

SC

(Jagged1, Jagged2, Delta-like 1(Dll-1), Dll-3, and Dll-4) have been accounted for to date [68, 70]. Binding of Notch ligand to an adjacent Notch receptor activates Notch signaling

NU

prompting the cleavage of Notch through a cascade of proteolytic cleavages by the

MA

metalloprotease, tumor necrosis factor-α-converting enzyme (TACE) and γ-secretase complexes [68, 71]. The cleavage by TACE generates Notch extracellular truncation

D

(NEXT) which is subsequently cleaved by the γ-secretase complex releasing the active

TE

fragment Notch intracellular domain (NICD) from the plasma membrane. NICD translocates into the nucleus where it binds to members of the CSL transcription factor

AC CE P

family and activates Notch target genes [68, 72]. Impaired Notch signaling is well reported in pancreatic cancers. Recently, it has been reported that inhibition of Notch signaling pathway by Notch1 siRNA or gammasecretase inhibitors, such as, MRK-003, MK-0752 etc., enhances chemosensitivity to gemcitabine in pancreatic cancer cells through activating apoptosis activity [73]. Abel et al. reported the significance of Notch pathway in maintaining cancer stem cell population in pancreatic cancer and investigated the connection of Notch pathway and percentage of the CSCs population. The inhibition of this pathway resulted in a reduced percentage of CSCs and tumor sphere formation; notwithstanding, activation demonstrated the inverse impact [74]. Further, Lee et al. suggested that the activation of the Notch pathway and the increase in CSCs might contribute to the failure of treatment in pancreatic cancer [75].

ACCEPTED MANUSCRIPT Notch has also been shown to be associated with the EMT in pancreatic cancer [72, 76]. It is remarkable that during the EMT process, epithelial cells gain the mesenchymal over

PT

endothelial characteristic thereby increasing in migratory and invasive capacity, leading

RI

to invasion and metastasis [77, 78].

SC

PI3K/Akt Pathway

NU

PI3K pathway acts via phosphorylation of FoxO proteins via Akt, which in turn impairs the DNA-binding ability and increases its affinity for 14-3-3 proteins [79]. These

MA

complexes are exported from the nucleus to cytoplasm leading to the inhibition of FoxOmediated survival pathways. Some other downstream effectors that regulate cell cycle

D

arrest and apoptosis, such as, active FKHRL1, FKHR, and AFX are also translocated to

TE

the cytoplasm by similar mechanism [79]. Downstream effectors of the PI3K-Akt

AC CE P

pathway are actively involved in a variety of vital and specialized functions such as differentiation and proliferation in diversified cells including adipocytes, hepatocytes, myoblasts, thymocytes and cancer cells [79]. PI3K/Akt pathway is activated in a variety of cells including fibroblastic, epithelial, and neuronal cells as survival signal. An elevated level of Akt has been reported in many types of tumors. Studies have shown the requirement of activated PI3K-Akt/FoxO signaling for the growth and survival of the pancreatic tumor. It has been found that the cells with elevated Akt levels are less sensitive to apoptosis stimuli. Akt regulates apoptosis directly by regulating its primary targets, Bad, and Caspase 9 and indirectly by controlling human telomerase reverse transcriptase subunit, FoxOs and IkappaB kinases and so forth [79].

ACCEPTED MANUSCRIPT Sonic Hedgehog Pathway Sonic hedgehog (Shh) is a member of the Hedgehog (Hh) family of secreted signaling

PT

proteins. Shh signaling is triggered by binding of the secreted Shh peptide to Patched

RI

(Ptch), which leads to inhibition of Ptch activity. Consequently, Smoothened (Smo) gets

SC

phosphorylated resulting in the activation of the Gli family of zinc-finger transcription factors and therefore target gene expression [80]. Shh signaling has diverse functions

NU

during vertebrate development and post-embryonically in tissue homeostasis [81, 82].

MA

Alteration in this pathway have been linked to various tumor types including pancreatic cancer [81, 82]. Activation of Shh signaling pathway has been reported to be involved in

D

the regulation of the pancreatic CSC's expansion, whereas its inhibition (by impairing Gli

TE

binding to its promoters) has been demonstrated to upregulate DRs and Fas expression, curb Bcl-2 and PDGFRα expressions, and encourage apoptotic cell death in pancreatic

AC CE P

CSCs [24]. Further, its inhibition has been shown to reduce tumor-associated stromal tissues, enhance gemcitabine uptake in tumor cells and prolong the average survival rate in pancreatic cancer mouse model [83]. Recently, Rodova et al. showed that inhibition of Shh pathway components, Gli transcriptional activity and its downstream targets by sulforaphane inhibited human pancreatic CSCs derived spheres and induced apoptosis by inhibition of Bcl-2 and activation of caspases in vitro [84]. Further, Li et al. showed that inhibition of Shh pathway by sulforaphane results in a marked reduction in EMT, metastatic, angiogenic markers with significant inhibition of tumor growth in mice. Since aberrant Shh signaling is frequently observed in pancreatic cancers, therapeutics that target Shh pathway and therefore CSCs, may improve the outcomes of patients with this devastating disease [85].

ACCEPTED MANUSCRIPT Treatment of Pancreatic Cancer: Chemotherapy and Radiotherapy The essential choice for pancreatic cancer treatment is its surgical removal. However, in

PT

advance metastatic stages, the treatment mainly aims to increase the survival by optimal

RI

control of metastases. Systemic chemotherapy may be utilized at any phase of pancreatic

SC

malignancy with the objective to minimize the patient’s disease-related symptoms and to prolong survival. Presently, a limited number of drugs are accessible for the treatment of

NU

the pancreatic tumor. Gemcitabine has been the reference regimen since 1997 when it

MA

was indicated better than 5-Fluorouracil (5-FU) in a phase III clinical trial [86]. A significant concern with 5-FU was the associated toxicity with its treatment, in particular,

D

gastrointestinal toxicity. A combination of 5-FU and Fluoropyrimidine (S-1) could

TE

likewise be a decent choice as S-1 potentiates the antitumor activity of 5-FU and decreases gastrointestinal toxicity in pancreatic tumor mouse models [87]. In recent years

AC CE P

different combinations, for example, the systemic treatment with Abraxane and Gemcitabine and the multidrug mix, FOLFIRINOX, have been attempted [88-109]. Despite the fact that FOLFIRINOX has been promising, it can have a remarkable reaction profile, constraining its utility in patients with poor baseline performance status [110]. Radiation therapy, combined with chemotherapy, may be used in patients whose cancers have grown beyond the pancreas and cannot be removed by surgery. Uses of radiation treatment alongside high-energy x-beams to kill cancer cells is extremely regular for the treatment of advanced stages of tumors. Pancreatic neuroendocrine tumors (NETs) usually do not respond to radiation, and therefore it is rarely used to treat these tumors. However, it can be used in case of pancreatic NETs that have spread to the bone and other tissues. Furthermore, the typical radiation treatment utilized for the treatment of the

ACCEPTED MANUSCRIPT exocrine pancreatic malignancies is External Shaft Radiation Treatment that focuses the radiation on the cancer from a machine outside the body. Radiation treatment is typically

PT

associated with different side effects, like skin changes in areas getting radiation, nausea

SC

but not the least increased risk of severe infection.

RI

and vomiting, diarrhea, fatigue, poor appetite, weight loss, lower blood counts and last

NU

Clinical Trials in Pancreatic Cancer

The chemotherapy drug, Gemcitabine, has been a standard initial treatment for patients

MA

with metastatic pancreatic cancer for over 15 years [86]. Various clinical trials have tried new medications, either alone or in combination with Gemcitabine (Table 1); however,

D

the advancement is moderate amid the most recent decade [88-109, 111]. Gemcitabine

TE

alone or in combination with Capecitabine or Erlotinib remained the favored systemic

AC CE P

treatment alternatives until 2010 [88-109]. Since 2010, use of FOLFIRINOX has increased both in metastatic and locally advanced cancer [110, 111]. Various drugs and combinations have been evaluated in the last couple of years, and some of them have shown promising results. Sunitinib and Everolimus have shown significant improvement in survival. Sunitinib treatment showed median progression-free survival of 11.4 months as compared with 5.5 months for patients who received the placebo. On the other hand, the patients receiving Everolimus showed median progression-free survival of 11 months as compared with 4.6 months for patients who received the placebo. Although these drugs have shown promising results, severe side effects as anemia and neutropenia were also observed.

ACCEPTED MANUSCRIPT Combination of Nab-Paclitaxel (Abraxane®), a form of the chemotherapy drug Paclitaxel bound to the human protein albumin and contained in nanoparticles, and Gemcitabine

PT

(Gemzar®) was also investigated in an international randomized phase III trial showing

RI

improved survival [112]. Patients who received the drug combination had a median overall survival of 8.5 months, compared with 6.7 months for patients treated with

SC

gemcitabine alone [112]. FDA approved therapy to treat patients with metastatic

NU

pancreatic cancer based on the results of the MPACT trial. Some other combinations, such as, gemcitabine with gamma-secretase inhibitor (MK-0752) or FG-3019 (a human

MA

monoclonal antibody that suppresses connective tissue growth factor), have also shown

D

promising results [113].

TE

Herbal products, cancer prevention, and pancreatic cancer

AC CE P

The relation between the consumption of certain dietary herbals and a reduced risk of cancer is becoming evident as many epidemiological and pre-clinical studies have shown the effect of herbals on health [114]. In the past few years, the cancer chemoprevention approach is directed towards polyphenols and their health-related properties and a wide range of dietary constituents show potential biological activities [114]. Studies in this line on cell/animal models and human epidemiological trials have shown the potential of dietary polyphenols as anti-carcinogenic agents. The reports have shown that phenolic compounds have the capability to inhibit the molecular events in the cancer initiation, promotion, and progression stages. They may increase the expression of pro-apoptotic components in initiated proliferating cells and thereby prevent or delay tumor development. Although it seems that phenolic compounds induce apoptosis in a precise manner in cancer cells but in some human studies no promising results were obtained. A

ACCEPTED MANUSCRIPT Cohort Study of Diet and Cancer in Netherlands suggested no effect of the consumption of black tea on the risk for colorectal, stomach, lung and breast cancers [115]. A similar

PT

study was performed in Japan involving more than 25,000 stomach cancer patients with a

RI

similar observation i.e. no association of consumption of green tea with gastric cancer risk [116]. Some other studies on different types of cancer also indicated the same

SC

conclusion [117-119]. On the contrary, a decreased risk for the different types of cancer

MA

these phenolic compounds [118-122].

NU

has been reported after the consumption of flavonoids or certain foods or drinks rich in

The use of herbals in the treatment of pancreatic cancer is a novel approach and is

D

continuously gaining the attention of investigators. Previous research in this area was

TE

focused on inducing apoptosis but recently these herbals have been used in the targeting of other key pathways of cell survival, angiogenesis, metastasis, and differentiation. Easy

AC CE P

availability and less or no toxicity even at higher doses have made them the preferable choice over other cancer chemopreventive options (Fig. 2). Resveratrol

Resveratrol, a phytoalexin, is commonly found as an ingredient in red wine, skins of grapes, peanuts and so forth. It possess anti-tumorigenic, anti-inflammatory, and antioxidant properties [123]. Its preventive role against cancers, cardiovascular diseases, and various neurological disorders has been widely reported [124-126]. Hydroxylation of resveratrol by CYP1B1 generates two major metabolites namely piceatannol and 3,4,5,4’-tetrahydroxystilbene that substantially contribute to its chemopreventive activities by inhibiting tyrosine kinase and inducing apoptosis [127-129].

ACCEPTED MANUSCRIPT Chemopreventive effects of resveratrol against various cancers have been extensively investigated in both in vitro and in vivo. It has been shown to have anti-tumor activity by

PT

inhibiting angiogenesis, endothelial cell migration, tumor formation and by blockage of

RI

oxygen free radical formation [130-132] (Table 2). Due to its lipophilic nature, it readily crosses the plasma membrane and establishes dynamic homeostasis by inhibiting the

SC

phase I (mainly CYP450s) and inducing phase II enzymes (UDP-glucuronosyl

NU

transferase, NAD(P)H quinone oxidoreductase, and glutathione-s-transferases) during stress conditions [133-138]. In cancerous cells, it inhibits the expression of inducible

MA

nitric oxide (NO) synthase and NO production [139]. It also inhibits the formation of a preneoplastic lesion in mouse mammary glands and proliferation of a variety of cancer

TE

D

cells in culture including, human colon, breast, and prostate cancer cells [140-143]. Resveratrol sensitizes a broad spectrum of tumors including lung carcinoma, acute

AC CE P

myeloid leukemia, promyelocytic leukemia, multiple myeloma, prostate cancer, oral epidermoid carcinoma, and pancreatic cancer.

Zhou et al. showed that it enhances

caspase-3 activation and p53 and p21 expression in capan-2 and colo357 pancreatic cancer cell lines [144]. A recent approach, using human pancreatic CSCs (CD133+CD44+CD24+ESA+), showed that resveratrol sensitizes and inhibits the growth and development of pancreatic cancer lesion in KrasG12D mice [10]. This study further showed that the resveratrol inhibits pluripotency maintaining factors (Nanog, Sox-2, cMyc and Oct-4), drug resistance gene ABCG2, CSC's migration, invasion, self-renewal, and components of EMT (Zeb-1, Slug, and Snail) [10]. Resveratrol inhibits cell growth, proliferation and expression of the anti-apoptotic proteins Bcl-2, Bcl-xL, and XIAP and induces apoptosis, cell cycle arrests, caspases and pro-apoptotic gene Bax in pancreatic

ACCEPTED MANUSCRIPT cancer cell lines [145]. Additionally, resveratrol has been found to suppress proliferation and anchorage-independent growth of pancreatic cancer by inhibiting leukotriene B4

PT

(LTB4) production and expression of the LTB4 receptor 1 (LTB4R1) [146]. It is

RI

remarkable that LTB4 is a hydrolysis product of the leukotriene A4 (LTA4), and the process is catalyzed by LTA4 hydrolase, a known target for prevention and therapy of

SC

cancers including PC [147]. Resveratrol can directly bind to leukotriene A4 hydrolase

NU

(LTA-4H) and inhibit its activity and, therefore, LTB4 production [146].

MA

Curcumin

Curcumin is one of the most commonly used and highly investigated phytochemical.

D

During the last decade, our understanding of its therapeutic potential and the multiple

TE

mechanisms by which it offers chemoprevention against various cancers has been

AC CE P

increased [148] (Table 3). The various pharmacological effects of curcumin include apoptotic, anti-proliferative, anti-oxidant, and anti-angiogenic properties. The previous studies have highlighted that curcumin targets multiple signal transduction pathways and that it suppresses a number of essential elements in cellular signaling pathways, for example, phosphorylation catalyzed by protein kinases, c-Jun-activated protein 1 (AP-1) activation and prostaglandin biosynthesis. It has also been found that curcumin potentiates radiotherapy in PC cure probably by involving selective regulation of radiotherapy-induced NF-κB [149]. Studies have shown that curcumin inhibits cell proliferation and induces apoptotic cell death mediated by PARP cleavage and Caspase-3 in MIAPaCa-2, Panc-1 and BxPC-3 pancreatic cancer cells [150]. Subramaniam et al. showed a significant reduction in tumor volume and

ACCEPTED MANUSCRIPT angiogenesis in curcumin treated tumor xenografts [151]. They further showed that curcumin inhibits cell proliferation, induces of G2-M arrest and apoptosis, enhances

PT

phosphorylation of checkpoint kinase 2 (Chk2) coupled with higher levels of nuclear

RI

cyclin B1 and Cdc-2, and increases expression of cyclooxygenase-2 (COX-2) [151]. Curcumin also inhibits ERK activity and suppresses EGFR and Notch-1 signaling leading

SC

to increased apoptosis in pancreatic cancer. Glienke et al. showed that incubation with

NU

curcumin results in down-regulation of Wilms' tumor gene 1 (WT1; a gene frequently expressed in pancreatic cancer) in a dose-dependent manner [152]. Additionally,

MA

curcumin has been shown to restrain STAT3 and induce apoptosis by inhibiting the

D

expression of the anti-apoptotic gene Survivin/BIRC4 in pancreatic cancer cells [153].

TE

Recently Bar-Sela et al. reviewed the accomplished and continuing clinical trials with curcumin as an anticancer agent [154]. In one trial, 17 patients were treated with the oral

AC CE P

dose of 8 gm/day of curcumin in combination with Gemcitabine. Although the results showed that this combined treatment is tolerable in patients; nevertheless, it has been suggested to reduce the dose of curcumin [155]. Dhillon et al. used only curcumin as the 1st line treatment for the 25 patients. They found that curcumin down regulates the expression of NFκB, COX-2, and the phosphorylation of STAT3 in peripheral blood [156]. In spite of these encouraging results, extensive clinical trials are needed before drawing any conclusion. Epigallocatechin gallate (EGCG) Epigallocatechin gallate (EGCG) is a most extensively studied catechin and the major polyphenol present in green tea. Various studies have shown that EGCG offers protection

ACCEPTED MANUSCRIPT against pancreatic cancer among the other tumors (Table 4); however, the exact molecular mechanism by which EGCG suppresses human pancreatic cancer cell

PT

proliferation is unclear. Kürbitz et al. showed anticancer properties of EGCG on human

RI

pancreatic ductal adenocarcinoma (PDAC) cells PancTu-I, Panc1, Panc89 and BxPC3 in vitro [157]. They found that EGCG inhibits proliferation of PDAC cells in a dose- and

SC

time-dependent manner. The protein expression analysis performed with PancTu-I cells

NU

evidently showed EGCG-mediated modulation of cell cycle regulatory proteins (cyclins, cyclin-dependent kinases, and inhibitors). The study further stated that EGCG inhibits

MA

TNFα-induced activation of NF-κB and consequently secretion of pro-inflammatory and invasion-promoting proteins like IL-8 and uPA [157]. Moreover, previous studies have

TE

D

demonstrated that EGCG decreases cell adhesion ability on micro-pattern dots, accompanied by dephosphorylation of both focal adhesion kinase and insulin-like growth

AC CE P

factor-1 receptor (IGF-1R) in AsPC-1 and BxPC-3 cells [158] [159] [64]. EGCG has been also found to aid retained activation of MAPK signaling, suppressed growth, reduced cell viability, and increased apoptosis in these cells in a dose-dependent manner [158].

Effects of EGCG on heat shock proteins were also investigated. A study by Li Y et al. showed that the binding of EGCG to Hsp90 impairs the association of Hsp90 with its cochaperones, thereby inducing degradation of Hsp90 client proteins (Akt, Cdk4, Raf-1, Her-2, and pERK) consequently anti-proliferating effects in pancreatic cancer cells [160]. Basu and Haldar showed that EGCG causes the disappearance of intact 21 kDa Bid protein and induces activation of caspase-8 leading to cell death in MIA PaCa-2 cells [161]. Further, involvement of transmembrane extrinsic signaling in this polyphenol

ACCEPTED MANUSCRIPT triggered pancreatic carcinoma cell death was confirmed by RNase protection assay that clearly showed up-regulation of the members of death receptor family [161] [160].

PT

Shankar et al. examined the role of EGCG in inhibiting growth, invasion, metastasis and

RI

angiogenesis of human pancreatic cancer cells in a xenograft model system [162]. They

SC

found that EGCG inhibits viability, capillary tube formation, and migration of HUVEC. Additionally, they observed EGCG-mediated inhibition of proliferation (Ki-67 and

NU

PCNA staining), angiogenesis (vWF, VEGF and CD31) and metastasis (MMP-2, MMP-

MA

7, MMP-9 and MMP-12), and induction of apoptosis (TUNEL), caspase-3 activity and growth arrest (p21/WAF1) in vivo [162]. They also found a significant reduction in the

D

circulating vascular endothelial growth factor receptor 2 (VEGF-R2) positive endothelial

Genistein

AC CE P

treatment [162].

TE

cells, ERK activity, and induction of p38 and JNK activities in vivo following EGCG

Genistein is found in a number of plants including lupin, fava beans, soybeans, kudzu, and psoralea, in the medicinal plant, Flemingia vestita, and coffee [159, 160, 163-165]. It has multiple effects in living cells, such as activation of PPARs, estrogen receptor-β, Nrf2 anti-oxidative response, stimulation of autophagy and inhibition of several tyrosine kinases, topoisomerase, and mammalian hexose transporter GLUT-1 [73, 166-174]. Genistein also affects tumor formation, cell multiplication and differentiation, angiogenesis, and signaling triggered by growth factors [175-184]. The most critical activity that contributes to the chemopreventive potential of genistein is tyrosine kinase inhibition, mostly of epidermal growth factor receptor EGFR. Additionally, the inhibitory

ACCEPTED MANUSCRIPT effect of genistein on DNA topoisomerase II is also a major contributor to its cytotoxic activity [170, 172].

PT

Genistein inhibits cell growth, clonogenicity, cell migration and invasion, EMT

RI

phenotype, and formation of pancreatospheres consistent with reduced expression of

SC

CD44 and EpCAM [185]. Wang et al., showed that genistein restricts pancreatic cancer cell invasion by inhibiting cell growth and inducing apoptosis along with attenuation of

NU

FoxM1 and its downstream genes (survivin, Cdc25a, MMP-9, and VEGF) [186].

MA

Sulforaphane

Sulforaphane has been reported to inhibit the growth of established tumors and prevent

TE

D

chemically induced cancers in animal models [187-189] (Table 5). It has been shown to inhibit Akt pathway in ovarian, prostate and colorectal cancers [189-191] and down-

AC CE P

regulate β-catenin in HeLa and HepG2 cells [192]. Additionally, it also targets breast cancer stem/progenitor cells effectively in both in vitro and in vivo conditions [193]. The studies have shown that recipient NOD/SCID mice inoculated with tumor cells derived from sulforaphane-treated primary xenograft failed to develop tumor growth, whereas control tumor cells quickly generate large tumors [194]. Furthermore, sulforaphane has also been shown to inhibit the self-renewal capacity of pancreatic CSCs. Srivastava et al., showed that inhibition of Nanog enhances the inhibitory effects of sulforaphane on the self-renewal capacity of CSCs [195]. Sulforaphane induces apoptosis by activating caspase-3 and inhibiting the expression of Bcl-2 and XIAP, as well as phosphorylation of FKHR. Additionally, sulforaphane is suggested to block signaling involved in early metastasis by inhibiting the expression of

ACCEPTED MANUSCRIPT proteins involved in the epithelial-mesenchymal transition (beta-catenin, vimentin, TWIST1, and ZEB1) [195]. Additionally, sulforaphane, in combination of with TRAIL,

PT

has been suggested to be a promising strategy for targeting pancreatic tumor initiating

RI

cells (TICs). It has been found that it could abrogate the resistance of pancreatic TICs to TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) by interfering with

NU

Garlic

SC

TRAIL-activated NF-κB signaling [196].

MA

Garlic has been traditionally used for varied human ailments around the world. Epidemiological observations and preclinical studies, both in cell and animal models,

D

suggest the anti-carcinogenic potential of garlic and its constituents [197]. Chemical

TE

analysis revealed that the protective effects of garlic are due to the presence of

AC CE P

organosulfur compounds mainly allyl derivatives [197]. Additionally, it modulates the activity of several metabolizing enzymes involved in the activation and detoxification of carcinogens and inhibits DNA adduct formation. It possess anti-oxidative and free radicals scavenging properties and regulates cell proliferation, apoptosis, and immune responses. Recent data suggest that garlic also modulates cell-signaling pathways to avoid proliferation of unwanted cells thereby imparting strong cancer chemopreventive, as well as cancer therapeutic effects [197]. Benzyl Isothiocyanate (BITC) Due to their capability to induce apoptosis, modulate signaling pathways and inhibit angiogenesis, isothiocyanates (ITCs) have shown a great promise as chemopreventive agents against various tumors in recent years [198]. Benzyl isothiocyanate (BITC) is a

ACCEPTED MANUSCRIPT major ITC compound present in cruciferous vegetables. BITC suppresses the initiation and progression of a variety of cancers including lung, esophageal, forestomach, urinary

PT

bladder, mammary, liver, colon, and pancreatic tumors [198-203]. Various preclinical

RI

and mechanistic studies have supported the anticancer efficacy of BITC, and it has been found to suppress the growth of human pancreatic cancer cells both in vitro and in vivo.

SC

BITC is reported to induce G(2)/M phase cell cycle arrest, and apoptotic cell death in

NU

pancreatic cancer cell/animal models [204-207]. The apoptotic potential of BITC is attributed to its capability to activate MAPK family members i.e. ERK, JNK and P38 by

MA

catalyzing their phosphorylation at Thr202/Tyr204, Thr183/Tyr185, and Thr180/Tyr182 respectively in a dose-dependent manner [206]. Additionally, the potential to inhibit the

TE

D

phosphorylation and expression of NF-kB most likely via inhibition of HDAC1/HDAC3, is suggested to be another contributory factor to the apoptotic potential of BITC [204].

AC CE P

Furthermore, it has been found to inhibit angiogenesis and metastasis by suppressing VEGF and MMP-2 expression in pancreatic cancer cells [208]. Recently Boreddy et al., showed that BITC offers protection against pancreatic tumor growth by effectively containing STAT-3 and HIF-1α and VEGF expression in BxPC-3 and PanC-1 pancreatic cancer cells [208]. It reduces the phosphorylation of PI3K, Akt, Pdk1, mTOR, FoxO1, and FoxO3a and increases apoptosis in tumor xenograft mouse model. BITC treatment also decreases the binding of FoxO1 with 14-3-3 protein suggesting it’s nuclear retention and subsequent elevation of FoxO-responsive proteins involved in apoptosis (Bim) and cell cycle arrest (p27 and p21) [209]. BITC has also been shown to sensitize pancreatic tumors for radiotherapy. BITC treatment in a combination of X-rays or gamma-irradiation reduces cell survival as

ACCEPTED MANUSCRIPT compared with individual (X-rays or gamma-irradiation) exposure in pancreatic cancer cells. This effect is suggested to be due to the inhibition of cell proliferation and anti-

PT

apoptotic genes like XIAP/IAP, and augmentation of apoptosis protease activating factor-

RI

1 (Apaf-1) triggered by BITC [208]. It is remarkable that Apaf-1 is essential for activation of caspase-9 in stress-induced apoptosis [208].

SC

Piperlongumine:

NU

Piperlongumine (PL) is an alkaloid found in the fruits of long pepper plants that displays

MA

potent growth-inhibitory properties in a variety of cancer cell lines and various animal models. It has been identified to target cancer cells selectively over normal cells through

D

an ROS-dependent mechanism in a cell-based small-molecule screening and quantitative

TE

proteomics approach [210]. It increases ROS levels and cancer-selective cell death by directly binding and inhibiting the antioxidant enzyme glutathione S-transferase pi 1

AC CE P

(GSTP1) [210, 211]. Raj et al., have shown that PL selectively targets pancreatic cancer cells, PANC-1, and MIA PaCa-2- both of which harbor mutated K-ras [210]. A recent study by Dhillon et al. further supported this finding and suggested that PL also targets BxPC-3 pancreatic cancer cells that contain wild-type K-ras [212]. They further showed the anti-cancer effects of PL in vivo. PL reduces tumor volume, increases oxidative DNA damage (8-OHdGhigh), and reduces proliferation (Ki-67low) in nude mice xenografts for PANC-1 [212]. Conclusion and future perspectives Pancreatic cancer is continuously posing a challenge to the clinicians and researchers. We are still relying on the old traditional therapies. The major drawback of current therapy is

ACCEPTED MANUSCRIPT their unilateral actions on one or two pathways whereas the approach should be to target several targets simultaneously. The combination therapy is a right approach in this

PT

direction, but associated side effects are a major concern. Since the small population of

RI

pancreatic CSCs is mostly responsible for the pathogenesis of pancreatic cancer, an efficient, targeted therapy for pancreatic CSCs is also an excellent approach. However,

SC

the problem is the resistance of pancreatic CSCs against conventional treatment but still

NU

developmental pathways such as the hedgehog, Wnt, Notch, etc. can be targeted.

MA

Flavonoids have emerged as potential chemopreventive candidates for cancer treatment, especially by their ability to induce apoptosis. These can interfere with the initiation,

D

development and progression of cancer by the modulation of cellular proliferation,

TE

differentiation, apoptosis, angiogenesis, and metastasis. Flavonoids have been shown to target cancer cells specifically with no or insignificant effects on healthy cells in vitro.

AC CE P

Nevertheless, some studies suggest to include experimental conditions (dose, cell type, culture conditions and treatment length) while interpreting the results of in vitro studies because the biological outcome can be affected. Since the apparent phenomenon is a result of complex interaction of different cellular events, the mechanisms for inducing the apoptosis of these polyphenols may overlap with other signaling cascades. Therefore, the promising strategy could be the promotion of programmed cell death through the modulation of different proteins in other pathways that can contribute to cell death. Flavonoids exhibit some characteristic effects, for example, induction of apoptosis, activation of caspases, down-regulation or up-regulation of Bcl-2 family members, induction of cell cycle arrest and inhibition of survival/proliferation signals. Moreover, the effects of these plant products on pancreatic cancer initiation, promotion and

ACCEPTED MANUSCRIPT metastasis has also discussed in the light of available literature. The results from in vitro experiments constitute a valuable tool for elucidating the pathways involved in the

PT

overall carcinogenesis process, although these cannot be directly extrapolated to clinical

RI

effects. The best thing about herbal products is that they target multiple signaling events simultaneously; however; more studies are needed to understand clearly the mechanisms

SC

of action of flavonoids as modulators of cell survival and apoptosis.

NU

Acknowledgments

MA

We acknowledge our lab members for critical reading of the manuscript, insightful discussions, and valuable advice. The project was funded by the National Institutes of

TE

D

Health (RKS) and The VA Merit Award (SS).

AC CE P

Table 1: List of some of the completed clinical trials on pancreatic cancer (Source: www. ClinicalTrials.gov) Rank

Interventions

NCT Number

1 2

Drug: RAD001 Drug: Gemcitabine|Drug: Albuminbound paclitaxel Drug: gemcitabine hydrochloride|Drug: imatinib mesylate Dietary Supplement: genistein|Drug: erlotinib hydrochloride|Drug: gemcitabine hydrochloride Drug: Ixabepilone|Drug: Cetuximab Biological: cetuximab|Drug: docetaxel|Drug: irinotecan hydrochloride Drug: alvocidib|Drug: docetaxel

Drug: bortezomib|Drug: carboplatin|Other: laboratory

3

4

5 6

7

8

Conditions

NCT00409292 NCT00398086

Enroll ment 33 67

NCT00161213

44

Pancreatic Cancer

NCT00376948

20

Pancreatic Cancer

NCT00383149 NCT00042939

58 94

Metastatic Pancreatic Cancer Pancreatic Cancer

NCT00331682

10

NCT00416793

9

Adenocarcinoma of the Pancreas|Recurrent Pancreatic Cancer|Stage IV Pancreatic Cancer Acinar Cell Adenocarcinoma of the Pancreas|Duct Cell

Pancreatic Cancer Metastatic Pancreatic Cancer

ACCEPTED MANUSCRIPT

Drug: bevacizumab [Avastin] Drug: Erlotinib, escalating dose|Drug: Erlotinib, standard dose|Drug: Gemcitabine

NCT01214720 NCT00652366

607 467

12

Drug: Ipilimumab|Biological: Pancreatic Cancer Vaccine Biological: cixutumumab|Drug: erlotinib hydrochloride|Drug: gemcitabine hydrochloride Drug: Gemcitabine|Drug: Erlotinib|Drug: Sorafenib Biological: bevacizumab|Drug: erlotinib hydrochloride|Other: laboratory biomarker analysis Drug: Capecitabine|Drug: Docetaxel Drug: Abraxane Drug: gamma-secretase/Notch signaling pathway inhibitor RO4929097

NCT00836407

30

NCT00617708

134

19 20

21 22 23

24

25

26 27 28

29

SC

Pancreatic Cancer

NCT00365144

36

Pancreatic Cancer

NCT00290693 NCT00691054 NCT01232829

45 20 18

NU

45

Drug: Lenalidomide|Drug: Gemcitabine Drug: sorafenib tosylate|Drug: gemcitabine hydrochloride|Other: laboratory biomarker analysis Drug: GSK1120212|Drug: Gemcitabine|Drug: Placebo Drug: Gemcitabine|Drug: AG013736|Drug: Gemcitabine Drug: dasatinib|Procedure: laboratory biomarker analysis|Procedure: physiologic testing Drug: gemcitabine|Drug: placebo|Drug: Erlotinib|Drug: apricoxib Drug: AG-013736|Drug: Gemcitabine|Drug: Gemcitabine|Drug: placebo Drug: Albumin-bound paclitaxel|Drug: Gemcitabine Drug: cohort 1|Drug: cohort 2|Drug: cohort 3|Drug: cohort 4 Drug: gemcitabine hydrochloride|Drug: tanespimycin

NCT00837031

72

Pancreatic Cancer Pancreatic Cancer Adenocarcinoma of the Pancreas|Recurrent Pancreatic Cancer|Stage IV Pancreatic Cancer Metastatic Pancreatic Cancer

NCT00114244

52

Stage IV Pancreatic Cancer

NCT01231581

160

Cancer

NCT00219557

111

Pancreatic Neoplasms

NCT00474812

49

NCT00709826

109

Adenocarcinoma of the Pancreas|Recurrent Pancreatic Cancer|Stage IV Pancreatic Cancer Pancreatic Cancer|Metastatic Pancreatic Cancer

NCT00471146

630

Carcinoma, Pancreatic Ductal

NCT00844649

861

Metastatic Pancreatic Cancer

NCT00439179

27

Metastatic Pancreatic Cancer

NCT00577889

21

Drug: PCI-27483|Drug: Gemcitabine

NCT01020006

42

Adenocarcinoma of the Pancreas|Recurrent Pancreatic Cancer|Stage IV Pancreatic Cancer Pancreatic Cancer|Ductal Adrenocarcinoma|Exocrine Pancreatic Cancer

MA

16 17 18

Stage IV Pancreatic Cancer

NCT00696696

D

15

TE

14

AC CE P

13

Pancreatic Cancer

RI

9 10

Adenocarcinoma of the Pancreas|Stage IV Pancreatic Cancer Pancreatic Cancer Pancreatic Cancer

PT

biomarker analysis

ACCEPTED MANUSCRIPT

37

38 39

41

42 43 44 45

Drug: Cetuximab|Drug: Gemcitabine|Drug: Oxaliplatin|Drug: Capecitabine|Radiation: Radiotherapy Drug: Gemcitabine

NCT00338039

69

Metastatic Pancreatic Adenocarcinoma Pancreatic Cancer

NCT00390182

38

Biological: DTA-H19 Drug: Fentanyl sublingual spray|Drug: Placebo Drug: Avastin|Drug: Tarceva|Radiation: Radiation Therapy Drug: Gemcitabine|Drug: Sunitinib

NCT00711997 NCT00538850

9 130

Gastrointestinal Neoplasms|Ovarian Neoplasms Pancreatic Neoplasms Cancer

NCT00735306

12

Pancreatic Cancer

NCT00556049

72

RI

PT

367

SC

36

NCT01124786

NU

34 35

Pancreatic Neoplasms

NCT00661830

103

Renal Cell Carcinoma|Neoplasm Metastases Adenocarcinoma

NCT00448136

83

Neoplasms

Drug: Avastin (Bevacizumab, RHUMAB VEGF)|Drug: Capecitabine|Radiation: Radiation Therapy Drug: Sunitinib

NCT00113230

25

Rectal Cancer

NCT01121562

12

Drug: RAD001|Drug: Octreotide Depot Drug: Sunitinib malate|Procedure: Hepatic Artery Embolizations Other: Cocoa Polyphenols

NCT00113360

67

NCT00434109

39

NCT01617603

62

Pancreatic Neuroendocrine Tumors Neuroendocrine Carcinoma|Islet Cell Carcinoma Neuroendocrine Tumor|Islet Cell Tumor Diabetes Type 2

Drug: Gemcitabine|Drug: Placebo|Drug: Sorafenib Drug: bevacizumab [Avastin]|Drug: 5 FU|Drug: Streptozotocin|Drug: Xeloda

MA

33

142

D

32

NCT00637247

TE

31

Drug: imexon in combination with gemcitabine|Drug: imexon placebo + gemcitabine Drug: CO-1.01|Drug: Gemcitabine

AC CE P

30

ACCEPTED MANUSCRIPT Table 2: Summary of recent findings showing chemopreventive potential of resveratrol against pancreatic cancer Effect

Reference

MIA PaCa-2 cells

Inhibits proliferation and induces apoptosis

Panc-28 and Hs766T

Increases calcium levels and prevents migration of TG2-

cells

expressing cells

Capan-2 cells

Inhibits tumor growth, induced apoptosis, and up-regulated Bax

[213]

[214]

MA

NU

SC

RI

PT

Cells

[215]

Inhibits cell proliferation, migration, and induces expression of

TE

BxPC-3 and Panc-1

D

and VEGF-B expression

[216]

EMT-related genes (E-cadherin, N-cadherin, vimentin, MMP-2,

PANC-1,

AC CE P

and MMP-9)

CFPAC-1,

Inhibits viability and miR-21 expression and increases Bcl2

[217]

and MIA Paca-2 cells

expression

BxPC-3 and Panc-1

Inhibits the growth Gli1, Ptc1, CCND1, and BCL-2

[218]

PANC-1, MIA PaCa-

Up-regulates p21/CIP1, p27/KIP1, Bim, activates csapase-3,

[219]

2, Hs766T, and AsPC-

reduces phosphorylation of ERK, PI3K, Akt, FoxO1, and

1

FoxO3a

ACCEPTED MANUSCRIPT Table 3: Summary of recent findings showing chemopreventive potential of curcumin against pancreatic cancer Effect

MIA PaCa-2

Inhibits the proliferation and enhances apoptosis in MIA PaCa-2 (tumor

xenograft model)

RI

mouse

[220]

cells and inhibits tumor growth and the expression of the transcription nuclear factor NF-κB and NF-κB-regulated gene

SC

and

Reference

PT

Cells/ animals

the

NU

products in xenograft mouse model MiaPaCa-2 and Panc-1

Down-regulates

expression

of

miR-221

resulting

in

cells

upregulation of PTEN, p27(kip1), p57(kip2), and PUMA leading

[221]

and

Inhibits cell proliferation, reduces tumor growth and angiogenesis

mouse

(tumor

xenograft model)

[222]

as determined by a reduced number of blood vessels and

D

MIA PaCa-2

MA

to the inhibition of cell proliferation and migration

decreased expression of vascular endothelial growth factor and

TE

annexin A2 proteins

Activates TNFR, CASP 8, CASP3, BID, BAX, and down-

2

regulates NFκB, NDRG 1, and BCL2L10 gene

AC CE P

BxPC-3 and MiaPaCa-

TGF-β1-stimulated

[223]

Inhibits proliferation, induces apoptosis and reverses the EMT

[224]

AsPC-1 and MiaPaCa-

Decreases

[225]

2

formation of pancreatospheres, invasive cell migration, and CSC

PANC-1 cells

pancreatic

cancer

cell

survival,

clonogenicity,

function AsPC-1,

MiaPaCa-2,

Panc-1, human

and

mouse cancer cells

Inhibits tumor growth through mitotic catastrophe by increasing

BxPC-3

the expression of RNA binding protein CUGBP2, thereby

Pan02

inhibiting the translation of COX-2 and VEGF expression

pancreatic

[151]

ACCEPTED MANUSCRIPT Table 4: Summary of recent findings showing chemopreventive potential of EGCG against pancreatic cancer Reference

PANC-1

Suppresses

proliferation

and

PT

Effect

induces

apoptosis,

RI

Cells/ animals

[226]

modulates the PI3K/Akt/mTOR signaling pathway Regulates RKIP/ERK/NF-κB and/or RKIP/NF-κB/Snail

SC

AsPC-1 cells

[227]

and inhibits invasive metastasis c

nude

mice

(tumor

xenograft model)

Inhibits pancreatic cancer orthotopic tumor growth,

NU

Balb

[162]

angiogenesis, and metastasis, inhibits PI3K/Akt and ERK

Colo357

human

pancreatic

adenocarcinoma cells

MA

pathways and activation of FKHRL1/FoxO3a With PGHS-2-specific inhibitor celecoxib, synergistically

[228]

diminishes metabolic activity via apoptosis induction and release

of

pro-angiogenic

vascular

D

down-regulates

TE

endothelial growth factor (VEGF) and invasiveness-

PANC-1

AC CE P

promoting matrix metalloproteinase (MMP)-2

Inhibits HIF-1α protein expression, P-gp mRNA and

[229]

protein levels and cell proliferation

AsPC-1 and PANC-1

Suppresses the growth, invasion, and migration, induces

[230]

apoptosis by interfering with the STAT3 signaling pathway and enhances the therapeutic potential of gemcitabine and CP690550

Human pancreatic cancer stem

Inhibits Nanog, c-Myc and Oct-4 expression, self-

cells

renewal, proliferation, EMT, components of Shh pathway

(CD133+/CD44+/CD24+/ESA+)

(smoothened, patched, Gli1 and Gli2) and Gli transcriptional activity, and induces apoptosis by inhibiting Bcl-2 and XIAP and activating caspase-3

[231]

ACCEPTED MANUSCRIPT Table 5: Summary of recent findings showing chemopreventive potential of sulforaphane against pancreatic cancer Effect

Established BxPc-3 and AsPC-1

Increases Cx43 and E-cadherin levels, inhibits c-Met and

PDA cell lines and immortalized

CD133, improved the functional morphology and

CRL-4023

communication of gap junctions.

human PDA cells

Inhibits cell viability and NF-κB DNA binding activity,

[233]

NU

MIA PaCa-2 and Panc-1

[232]

RI

SC

hTERT-HPNE

Reference

PT

Cells/ animals

induces cell apoptosis by activation of caspase-3 and PARP cleavage, increases pERK1/2, c-Jun, p38 MAPK,

MA

p53 protein expression when used in combination with aspirin and curcumin NOD/SCID/IL2Rgamma

mice

components,

D

(tumor xenograft model)

Inhibits growth of tumors, expression of Shh pathway EMT,

pluripotency

[85]

maintaining

TE

transcription factors, angiogenic markers and induces apoptosis Inhibits CSC’s derived spheres, components of Shh

cells

human

pathway and Gli transcriptional activity, expression of

pancreatic cancer stem cells

pluripotency maintaining factors (Nanog and Oct-4) as

(CD133+/CD44+/CD24+/ESA+)

well as PDGFRα and Cyclin D1

Pancreatic

Disrupts protein-protein interaction in Hsp90 complex for

AC CE P

Human normal pancreatic stem (HPSC)

and

cancer

mouse model

xenograft

[84]

[234]

its chemopreventive activity

MIA-PaCa2

Potentiates the inhibitory effects of gemcitabine and 5-

[235]

flurouracil on clonogenicity, spheroid formation, ALDH1 activity, Notch-1 and c-Rel expression

PANC-1, AsPC-1

MIA

PaCa-2

and

Inhibits cell proliferation, colony formation, phosphorylation of Akt and ERK, activates FoxO transcription factors and induces apoptosis

[79]

ACCEPTED MANUSCRIPT References

AC CE P

TE

D

MA

NU

SC

RI

PT

[1] A. Maitra, R. Ashfaq, C.R. Gunn, A. Rahman, C.J. Yeo, T.A. Sohn, J.L. Cameron, R.H. Hruban, R.E. Wilentz, Cyclooxygenase 2 expression in pancreatic adenocarcinoma and pancreatic intraepithelial neoplasia: an immunohistochemical analysis with automated cellular imaging, Am J Clin Pathol, 118 (2002) 194-201. [2] T. Furukawa, R. Fujisaki, Y. Yoshida, N. Kanai, M. Sunamura, T. Abe, K. Takeda, S. Matsuno, A. Horii, Distinct progression pathways involving the dysfunction of DUSP6/MKP-3 in pancreatic intraepithelial neoplasia and intraductal papillary-mucinous neoplasms of the pancreas, Mod Pathol, 18 (2005) 1034-1042. [3] Y. Tao, F. Lin, T. Li, J. Xie, C. Shen, Z. Zhu, Epigenetically Modified Pancreatic Carcinoma PANC-1 Cells Can Act as Cancer Vaccine to Enhance Antitumor Immune Response in Mice, Oncol Res, 21 (2013) 307-316. [4] S.M. Conrath, The use of epidemiology, scientific data, and regulatory authority to determine risk factors in cancers of some organs of the digestive system. 6. Pancreatic cancer, Regul Toxicol Pharmacol, 6 (1986) 193-210. [5] K.D. Lillemoe, C.J. Yeo, J.L. Cameron, Pancreatic cancer: state-of-the-art care, CA: a cancer journal for clinicians, 50 (2000) 241-268. [6] C. Pierantoni, A. Pagliacci, M. Scartozzi, R. Berardi, M. Bianconi, S. Cascinu, Pancreatic cancer: progress in cancer therapy, Critical reviews in oncology/hematology, 67 (2008) 27-38. [7] A. Neesse, S. Krug, T.M. Gress, D.A. Tuveson, P. Michl, Emerging concepts in pancreatic cancer medicine: targeting the tumor stroma, OncoTargets and therapy, 7 (2013) 33-43. [8] M. Bardou, I. Le Ray, Treatment of pancreatic cancer: A narrative review of costeffectiveness studies, Best practice & research. Clinical gastroenterology, 27 (2013) 881892. [9] T. Kolodecik, C. Shugrue, M. Ashat, E.C. Thrower, Risk factors for pancreatic cancer: underlying mechanisms and potential targets, Frontiers in physiology, 4 (2013) 415. [10] S. Shankar, D. Nall, S.N. Tang, D. Meeker, J. Passarini, J. Sharma, R.K. Srivastava, Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelialmesenchymal transition, PLoS One, 6 (2011) e16530. [11] U. Kapil, M. Srivastav, D. Nayar, Vitamin 'A' and oesophageal cancer, Trop Gastroenterol, 14 (1993) 87-90. [12] E.S. Kim, F.R. Khuri, Chemoprevention of lung cancer, Curr Oncol Rep, 4 (2002) 341-346. [13] B.L. Halvorsen, K. Holte, M.C. Myhrstad, I. Barikmo, E. Hvattum, S.F. Remberg, A.B. Wold, K. Haffner, H. Baugerod, L.F. Andersen, O. Moskaug, D.R. Jacobs, Jr., R. Blomhoff, A systematic screening of total antioxidants in dietary plants, J Nutr, 132 (2002) 461-471. [14] S. Gupta, H. Mukhtar, Chemoprevention of skin cancer: current status and future prospects, Cancer Metastasis Rev, 21 (2002) 363-380. [15] S. Gupta, Prostate cancer chemoprevention: current status and future prospects, Toxicol Appl Pharmacol, 224 (2007) 369-376.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[16] P.G. Pietta, Flavonoids as antioxidants, J Nat Prod, 63 (2000) 1035-1042. [17] B. Halliwell, J.M. Gutteridge, Biologically relevant metal ion-dependent hydroxyl radical generation. An update, FEBS Lett, 307 (1992) 108-112. [18] A.R. Silva, P.F. Menezes, T. Martinello, G.F. Novakovich, C.E. Praes, I.H. Feferman, Antioxidant kinetics of plant-derived substances and extracts, Int J Cosmet Sci, 32 (2010) 73-80. [19] M. Hidalgo, Pancreatic cancer, N Engl J Med, 362 (2010) 1605-1617. [20] M. Hockel, P. Vaupel, Tumor hypoxia: definitions and current clinical, biologic, and molecular aspects, J Natl Cancer Inst, 93 (2001) 266-276. [21] D. Shweiki, A. Itin, D. Soffer, E. Keshet, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis, Nature, 359 (1992) 843-845. [22] E.V. Abel, D.M. Simeone, Biology and clinical applications of pancreatic cancer stem cells, Gastroenterology, 144 (2013) 1241-1248. [23] C. Li, D.G. Heidt, P. Dalerba, C.F. Burant, L. Zhang, V. Adsay, M. Wicha, M.F. Clarke, D.M. Simeone, Identification of pancreatic cancer stem cells, Cancer research, 67 (2007) 1030-1037. [24] C.J. Lee, J. Dosch, D.M. Simeone, Pancreatic cancer stem cells, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 26 (2008) 2806-2812. [25] M. Herreros-Villanueva, A. Zubia-Olascoaga, L. Bujanda, c-Met in pancreatic cancer stem cells: therapeutic implications, World journal of gastroenterology : WJG, 18 (2012) 5321-5323. [26] P.C. Hermann, S.L. Huber, T. Herrler, A. Aicher, J.W. Ellwart, M. Guba, C.J. Bruns, C. Heeschen, Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer, Cell stem cell, 1 (2007) 313-323. [27] J. Fredebohm, M. Boettcher, C. Eisen, M.M. Gaida, A. Heller, S. Keleg, J. Tost, K.M. Greulich-Bode, A. Hotz-Wagenblatt, M. Lathrop, N.A. Giese, J.D. Hoheisel, Establishment and characterization of a highly tumourigenic and cancer stem cell enriched pancreatic cancer cell line as a well defined model system, PLoS One, 7 (2012) e48503. [28] A. Avan, K. Quint, F. Nicolini, N. Funel, A.E. Frampton, M. Maftouh, S. Pelliccioni, G.J. Schuurhuis, G.J. Peters, E. Giovannetti, Enhancement of the antiproliferative activity of gemcitabine by modulation of c-Met pathway in pancreatic cancer, Current pharmaceutical design, 19 (2013) 940-950. [29] G. Luo, J. Long, X. Cui, Z. Xiao, Z. Liu, S. Shi, L. Liu, C. Liu, J. Xu, M. Li, X. Yu, Highly lymphatic metastatic pancreatic cancer cells possess stem cell-like properties, International journal of oncology, 42 (2013) 979-984. [30] A. Van den Broeck, H. Vankelecom, W. Van Delm, L. Gremeaux, J. Wouters, J. Allemeersch, O. Govaere, T. Roskams, B. Topal, Human pancreatic cancer contains a side population expressing cancer stem cell-associated and prognostic genes, PLoS One, 8 (2013) e73968. [31] C.P. Tanase, A.I. Neagu, L.G. Necula, C. Mambet, A.M. Enciu, B. Calenic, M.L. Cruceru, R. Albulescu, Cancer stem cells: involvement in pancreatic cancer pathogenesis and perspectives on cancer therapeutics, World journal of gastroenterology : WJG, 20 (2014) 10790-10801.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[32] A. Amsterdam, C. Raanan, N. Polin, E. Melzer, D. Givol, L. Schreiber, Modulation of c-kit expression in pancreatic adenocarcinoma: a novel stem cell marker responsible for the progression of the disease, Acta histochemica, 116 (2014) 197-203. [33] J.A. Castellanos, N.B. Merchant, N.S. Nagathihalli, Emerging targets in pancreatic cancer: epithelial-mesenchymal transition and cancer stem cells, OncoTargets and therapy, 6 (2013) 1261-1267. [34] Y. Lu, H. Zhu, H. Shan, J. Lu, X. Chang, X. Li, J. Lu, X. Fan, S. Zhu, Y. Wang, Q. Guo, L. Wang, Y. Huang, M. Zhu, Z. Wang, Knockdown of Oct4 and Nanog expression inhibits the stemness of pancreatic cancer cells, Cancer letters, 340 (2013) 113-123. [35] S. Yabuuchi, S.G. Pai, N.R. Campbell, R.F. de Wilde, E. De Oliveira, P. Korangath, M.M. Streppel, Z.A. Rasheed, M. Hidalgo, A. Maitra, N.V. Rajeshkumar, Notch signaling pathway targeted therapy suppresses tumor progression and metastatic spread in pancreatic cancer, Cancer letters, 335 (2013) 41-51. [36] P. Maisonneuve, B.C. Marshall, A.B. Lowenfels, Risk of pancreatic cancer in patients with cystic fibrosis, Gut, 56 (2007) 1327-1328. [37] H. Yu, D. Pardoll, R. Jove, STATs in cancer inflammation and immunity: a leading role for STAT3, Nature reviews. Cancer, 9 (2009) 798-809. [38] A. Maitra, R.H. Hruban, Pancreatic cancer, Annu Rev Pathol, 3 (2008) 157-188. [39] M.P. di Magliano, C.D. Logsdon, Roles for KRAS in pancreatic tumor development and progression, Gastroenterology, 144 (2013) 1220-1229. [40] E. Rozenblum, M. Schutte, M. Goggins, S.A. Hahn, S. Panzer, M. Zahurak, S.N. Goodman, T.A. Sohn, R.H. Hruban, C.J. Yeo, S.E. Kern, Tumor-suppressive pathways in pancreatic carcinoma, Cancer research, 57 (1997) 1731-1734. [41] G. Feldmann, R. Beaty, R.H. Hruban, A. Maitra, Molecular genetics of pancreatic intraepithelial neoplasia, J Hepatobiliary Pancreat Surg, 14 (2007) 224-232. [42] A.H. Shain, J.R. Pollack, The spectrum of SWI/SNF mutations, ubiquitous in human cancers, PLoS One, 8 (2013) e55119. [43] S. Jones, X. Zhang, D.W. Parsons, J.C. Lin, R.J. Leary, P. Angenendt, P. Mankoo, H. Carter, H. Kamiyama, A. Jimeno, S.M. Hong, B. Fu, M.T. Lin, E.S. Calhoun, M. Kamiyama, K. Walter, T. Nikolskaya, Y. Nikolsky, J. Hartigan, D.R. Smith, M. Hidalgo, S.D. Leach, A.P. Klein, E.M. Jaffee, M. Goggins, A. Maitra, C. Iacobuzio-Donahue, J.R. Eshleman, S.E. Kern, R.H. Hruban, R. Karchin, N. Papadopoulos, G. Parmigiani, B. Vogelstein, V.E. Velculescu, K.W. Kinzler, Core signaling pathways in human pancreatic cancers revealed by global genomic analyses, Science, 321 (2008) 1801-1806. [44] C. Schindler, D.E. Levy, T. Decker, JAK-STAT signaling: from interferons to cytokines, J Biol Chem, 282 (2007) 20059-20063. [45] M. Bar-Natan, E.A. Nelson, M. Xiang, D.A. Frank, STAT signaling in the pathogenesis and treatment of myeloid malignancies, Jakstat, 1 (2012) 55-64. [46] P. Yue, J. Turkson, Targeting STAT3 in cancer: how successful are we?, Expert Opin Inv Drug, 18 (2009) 45-56. [47] C. Schindler, D.E. Levy, T. Decker, JAK-STAT signaling: from interferons to cytokines, J Biol Chem, 282 (2007) 20059-20063. [48] M. Bar-Natan, E.A. Nelson, M. Xiang, D.A. Frank, STAT signaling in the pathogenesis and treatment of myeloid malignancies, JAKSTAT, 1 55-64. [49] P. Yue, J. Turkson, Targeting STAT3 in cancer: how successful are we?, Expert Opin Investig Drugs, 18 (2009) 45-56.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[50] A.K. Mankan, F.R. Greten, Inhibiting signal transducer and activator of transcription 3: rationality and rationale design of inhibitors, Expert Opin Inv Drug, 20 (2011) 12631275. [51] M.S. Chang, H.S. Lee, B.L. Lee, Y.T. Kim, J.S. Lee, W.H. Kim, Differential protein expression between esophageal squamous cell carcinoma and dysplasia, and prognostic significance of protein markers, Pathol Res Pract, 201 (2005) 417-425. [52] A. Scholz, S. Heinze, K.M. Detjen, M. Peters, M. Welzel, P. Hauff, M. Schirner, B. Wiedenmann, S. Rosewicz, Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer, Gastroenterology, 125 (2003) 891-905. [53] T. Toyonaga, K. Nakano, M. Nagano, G. Zhao, K. Yamaguchi, S. Kuroki, T. Eguchi, K. Chijiiwa, M. Tsuneyoshi, M. Tanaka, Blockade of constitutively activated Janus kinase/signal transducer and activator of transcription-3 pathway inhibits growth of human pancreatic cancer, Cancer letters, 201 (2003) 107-116. [54] T. Miyatsuka, H. Kaneto, T. Shiraiwa, T.A. Matsuoka, K. Yamamoto, K. Kato, Y. Nakamura, S. Akira, K. Takeda, Y. Kajimoto, Y. Yamasaki, E.P. Sandgren, Y. Kawaguchi, C.V. Wright, Y. Fujitani, Persistent expression of PDX-1 in the pancreas causes acinar-to-ductal metaplasia through Stat3 activation, Genes Dev, 20 (2006) 14351440. [55] D. Wei, X. Le, L. Zheng, L. Wang, J.A. Frey, A.C. Gao, Z. Peng, S. Huang, H.Q. Xiong, J.L. Abbruzzese, K. Xie, Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis, Oncogene, 22 (2003) 319-329. [56] M. Lesina, M.U. Kurkowski, K. Ludes, S. Rose-John, M. Treiber, G. Kloppel, A. Yoshimura, W. Reindl, B. Sipos, S. Akira, R.M. Schmid, H. Algul, Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer, Cancer Cell, 19 (2011) 456-469. [57] Y.G. Chen, A.M. Meng, Negative regulation of TGF-beta signaling in development, Cell research, 14 (2004) 441-449. [58] Y. Shi, J. Massague, Mechanisms of TGF-beta signaling from cell membrane to the nucleus, Cell, 113 (2003) 685-700. [59] T. Jonson, E. Albrechtsson, J. Axelson, M. Heidenblad, L. Gorunova, B. Johansson, M. Hoglund, Altered expression of TGFB receptors and mitogenic effects of TGFB in pancreatic carcinomas, International journal of oncology, 19 (2001) 71-81. [60] T. Jonson, M. Heidenblad, P. Hakansson, L. Gorunova, B. Johansson, T. Fioretos, M. Hoglund, Pancreatic carcinoma cell lines with SMAD4 inactivation show distinct expression responses to TGFB1, Genes, chromosomes & cancer, 36 (2003) 340-352. [61] J.P.t. Morris, S.C. Wang, M. Hebrok, KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma, Nature reviews. Cancer, 10 (2010) 683-695. [62] K.R. Brennan, A.M. Brown, Wnt proteins in mammary development and cancer, J Mammary Gland Biol Neoplasia, 9 (2004) 119-131. [63] C.Y. Logan, R. Nusse, The Wnt signaling pathway in development and disease, Annu Rev Cell Dev Biol, 20 (2004) 781-810. [64] M.M. Al-Aynati, N. Radulovich, R.H. Riddell, M.S. Tsao, Epithelial-cadherin and beta-catenin expression changes in pancreatic intraepithelial neoplasia, Clinical cancer

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

research : an official journal of the American Association for Cancer Research, 10 (2004) 1235-1240. [65] G. Zeng, M. Germinaro, A. Micsenyi, N.K. Monga, A. Bell, A. Sood, V. Malhotra, N. Sood, V. Midda, D.K. Monga, D.M. Kokkinakis, S.P. Monga, Aberrant Wnt/betacatenin signaling in pancreatic adenocarcinoma, Neoplasia, 8 (2006) 279-289. [66] M. Pasca di Magliano, A.V. Biankin, P.W. Heiser, D.A. Cano, P.J. Gutierrez, T. Deramaudt, D. Segara, A.C. Dawson, J.G. Kench, S.M. Henshall, R.L. Sutherland, A. Dlugosz, A.K. Rustgi, M. Hebrok, Common activation of canonical Wnt signaling in pancreatic adenocarcinoma, PLoS One, 2 (2007) e1155. [67] B. Wang, Q. Zou, M. Sun, J. Chen, T. Wang, Y. Bai, Z. Chen, B. Chen, M. Zhou, Reversion of trichostatin A resistance via inhibition of the Wnt signaling pathway in human pancreatic cancer cells, Oncology reports, (2014). [68] Z. Wang, Y. Li, F.H. Sarkar, Notch signaling proteins: legitimate targets for cancer therapy, Current protein & peptide science, 11 (2010) 398-408. [69] Z. Wang, Y. Li, S. Banerjee, F.H. Sarkar, Exploitation of the Notch signaling pathway as a novel target for cancer therapy, Anticancer research, 28 (2008) 3621-3630. [70] L. Miele, H. Miao, B.J. Nickoloff, NOTCH signaling as a novel cancer therapeutic target, Current cancer drug targets, 6 (2006) 313-323. [71] M.E. Fortini, Notch signaling: the core pathway and its posttranslational regulation, Dev Cell, 16 (2009) 633-647. [72] J. Ma, J. Xia, L. Miele, F.H. Sarkar, Z. Wang, Notch Signaling Pathway in Pancreatic Cancer Progression, Pancreatic disorders & therapy, 3 (2013). [73] X. Du, Y.H. Wang, Z.Q. Wang, Z. Cheng, Y. Li, J.K. Hu, Z.X. Chen, Z.G. Zhou, [Down-regulation of Notch1 by small interfering RNA enhances chemosensitivity to gemcitabine in pancreatic cancer cells through activating apoptosis activity], Zhejiang Da Xue Xue Bao Yi Xue Ban, 43 (2014) 313-318. [74] E.V. Abel, E.J. Kim, J. Wu, M. Hynes, F. Bednar, E. Proctor, L. Wang, M.L. Dziubinski, D.M. Simeone, The Notch pathway is important in maintaining the cancer stem cell population in pancreatic cancer, PLoS One, 9 (2014) e91983. [75] J.Y. Lee, S.Y. Song, J.Y. Park, Notch pathway activation is associated with pancreatic cancer treatment failure, Pancreatology : official journal of the International Association of Pancreatology, 14 (2014) 48-53. [76] Z. Wang, Y. Li, D. Kong, F.H. Sarkar, The role of Notch signaling pathway in epithelial-mesenchymal transition (EMT) during development and tumor aggressiveness, Current drug targets, 11 (2010) 745-751. [77] B. De Craene, G. Berx, Regulatory networks defining EMT during cancer initiation and progression, Nature reviews. Cancer, 13 (2013) 97-110. [78] A.D. Rhim, E.T. Mirek, N.M. Aiello, A. Maitra, J.M. Bailey, F. McAllister, M. Reichert, G.L. Beatty, A.K. Rustgi, R.H. Vonderheide, S.D. Leach, B.Z. Stanger, EMT and dissemination precede pancreatic tumor formation, Cell, 148 (2012) 349-361. [79] S.K. Roy, R.K. Srivastava, S. Shankar, Inhibition of PI3K/AKT and MAPK/ERK pathways causes activation of FOXO transcription factor, leading to cell cycle arrest and apoptosis in pancreatic cancer, Journal of molecular signaling, 5 (2010) 10. [80] T. Osterlund, P. Kogerman, Hedgehog signalling: how to get from Smo to Ci and Gli, Trends in cell biology, 16 (2006) 176-180.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[81] M. Varjosalo, J. Taipale, Hedgehog: functions and mechanisms, Genes Dev, 22 (2008) 2454-2472. [82] P.A. Beachy, S.S. Karhadkar, D.M. Berman, Tissue repair and stem cell renewal in carcinogenesis, Nature, 432 (2004) 324-331. [83] G. Feldmann, N. Habbe, S. Dhara, S. Bisht, H. Alvarez, V. Fendrich, R. Beaty, M. Mullendore, C. Karikari, N. Bardeesy, M.M. Ouellette, W. Yu, A. Maitra, Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer, Gut, 57 (2008) 1420-1430. [84] M. Rodova, J. Fu, D.N. Watkins, R.K. Srivastava, S. Shankar, Sonic hedgehog signaling inhibition provides opportunities for targeted therapy by sulforaphane in regulating pancreatic cancer stem cell self-renewal, PLoS One, 7 (2012) e46083. [85] S.H. Li, J. Fu, D.N. Watkins, R.K. Srivastava, S. Shankar, Sulforaphane regulates self-renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehogGLI pathway, Molecular and cellular biochemistry, 373 (2013) 217-227. [86] H.A. Burris, 3rd, M.J. Moore, J. Andersen, M.R. Green, M.L. Rothenberg, M.R. Modiano, M.C. Cripps, R.K. Portenoy, A.M. Storniolo, P. Tarassoff, R. Nelson, F.A. Dorr, C.D. Stephens, D.D. Von Hoff, Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 15 (1997) 2403-2413. [87] M. Fukushima, [Antitumor activity and function of S-1, a new oral tegafur-based formulation], Gan to kagaku ryoho. Cancer & chemotherapy, 33 Suppl 1 (2006) 19-26. [88] G. Abou-Alfa, R. Letourneau, G. Harker, M. Modiano, H. Hurwitz, N. Tchekmedyian, K. Feit, J. Ackerman, R. De Jager, S. Eckhardt, E. O'Reilly, Randomized phase III study of exatecan and gemcitabine compared with gemcitabine alone in untreated advanced pancreatic cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 24 (2006) 4441 - 4447. [89] J. Berlin, P. Catalano, J. Thomas, J. Kugler, D. Haller, A. Benson, E.C.O.G.T. E, Phase III study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 20 (2002) 3270 - 3275. [90] S. Bramhall, J. Schulz, J. Nemunaitis, P. Brown, M. Baillet, J. Buckels, A doubleblind placebo-controlled, randomised study comparing gemcitabine and Marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer, Br J Cancer, 87 (2002) 161 - 167. [91] G. Colucci, R. Labianca, F. Di Costanzo, R. Labianca, F. Di Costanzo, V. Gebbia, G. Carteni, B. Massidda, E. Dapretto, L. Manzione, E. Piazza, M. Sannicolo, M. Ciaparrone, L. Cavanna, F. Giuliani, E. Maiello, A. Testa, P. Pederzoli, M. Falconi, C. Gallo, M. Di Maio, F. Perrone, Randomized phase III trial of gemcitabine plus cisplatin compared with single-agent gemcitabine as first-line treatment of patients with advanced pancreatic cancer: the GIP-1 study, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 28 (2010) 1645 - 1651. [92] D. Cunningham, I. Chau, D. Stocken, J. Valle, D. Smith, W. Steward, P. Harper, J. Dunn, C. Tudur-Smith, J. West, S. Falk, A. Crellin, F. Adab, J. Thompson, P. Leonard, J. Ostrowski, M. Eatock, W. Scheithauer, R. Herrmann, J. Neoptolemos, Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

with advanced pancreatic cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 27 (2009) 5513 - 5518. [93] V. Heinemann, D. Quietzsch, F. Gieseler, M. Gonnermann, H. Schonekas, A. Rost, H. Neuhaus, C. Haag, M. Clemens, B. Heinrich, U. Vehling-Kaiser, M. Fuchs, D. Fleckenstein, W. Gesierich, D. Uthgenannt, H. Einsele, A. Holstege, A. Hinke, A. Schalhorn, R. Wilkowski, Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 24 (2006) 3946 - 3952. [94] V. Heinemann, V.-K. Ursula, W. Dirk, E. Kettner, A. Marten, C. Winkelmann, S. Klein, G. Kojouharoff, T. Gauler, L. von Weikersthal, M. Clemens, M. Geissler, T. Greten, S. Hegewisch-Becker, O. Rubanov, G. Baake, T. Hohler, Y. Ko, A. Jung, S. Neugebauer, S. Boeck, Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the 'Arbeitsgemeinschaft Internistische Onkologie' (AIO-PK0104), Gut, 62 (2012) 751 - 759. [95] R. Herrmann, G. Bodoky, T. Ruhstaller, B. Glimelius, E. Bajetta, J. Schuller, P. Saletti, J. Bauer, A. Figer, B. Pestalozzi, C. Kohne, W. Mingrone, S. Stemmer, K. Tamas, G. Kornek, D. Koeberle, S. Cina, J. Bernhard, D. Dietrich, W. Scheithauer, Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase III trial of the swiss group for clinical cancer Res and the central european cooperative oncology group, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 25 (2007) 2212 - 2217. [96] H. Kindler, T. Ioka, D. Richel, J. Bennouna, R. Letourneau, T. Okusaka, A. Funakoshi, J. Furuse, Y. Park, S. Ohkawa, G. Springett, H. Wasan, P. Trask, P. Bycott, A. Ricart, S. Kim, E. Van Cutsem, Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: a double-blind randomised phase 3 study, Lancet Oncol, 12 (2011) 25. [97] C. Louvet, R. Labianca, P. Hammel, G. Lledo, M. Zampino, T. Andre, A. Zaniboni, M. Ducreux, E. Aitini, J. Taieb, R. Faroux, C. Lepere, A. de Gramont, GERCOR, GISCAD, Gemcitabine in combination with Oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 23 (2005) 3509 - 3516. [98] M. Moore, D. Goldstein, J. Hamm, A. Figer, J. Hecht, S. Gallinger, H. Au, P. Murawa, D. Walde, R. Wolff, D. Campos, R. Lim, K. Ding, G. Clark, T. VoskoglouNomikos, M. Ptasynski, W. Parulekar, Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 25 (2007) 1960 - 1966. [99] H. Oettle, D. Richards, R. Ramanathan, J. van Laethem, M. Peeters, M. Fuchs, A. Zimmermann, W. John, D. Von Hoff, M. Arning, H. Kindler, A phase III trial of pemetrexed plus gemcitabine versus gemcitabine in patients with unresectable or metastatic pancreatic cancer, Ann Oncol, 16 (2005) 1639 - 1645. [100] P. Philip, J. Benedetti, C. Corless, R. Wong, E. O'Reilly, P. Flynn, K. Rowland, J. Atkins, B. Mirtsching, S. Rivkin, A. Khorana, B. Goldman, C. Fenoglio-Preiser, J.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

Abbruzzese, C. Blanke, Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group- directed intergroup trial S0205, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 28 (2010) 3605 - 3610. [101] E. Poplin, D. Levy, J. Berlin, H. Ochster, E. Mitchell, S. Alberts, P. O'Dwyer, D. Haller, P. Catalano, D. Cella, A. Benson, Phase III trial of gemcitabine (30-minute infusion) versus gemcitabine (fixed-dose-rate infusion [FDR]) versus gemcitabine oxaliplatin (GEMOX) in patients with advanced pancreatic cancer (E6201), Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 24 (2006) 933S. [102] M. Reni, S. Cereda, P. Passoni, A. Rognone, E. Mazza, R. Nicoletti, P. Arcidiacono, A. Zerbi, G. Balzano, V. Di Carlo, A randomized phase II trial of PEXG (cisplatin, epirubicin, capecitabine, gemcitabine) or PDXG (docetaxel) regimen in advanced pancreatic adenocarcinoma, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 25 (2007) A4628. [103] M. Reni, S. Cordio, C. Milandri, P. Passoni, P. Passoni, E. Bonetto, C. Oliani, G. Luppi, R. Nicoletti, L. Galli, R. Bordonaro, A. Passardi, A. Zerbi, G. Balzano, L. Aldrighetti, C. Staudacher, E. Villa, V. Di Carlo, Gemcitabine versus cisplatin, epirubicin, fluorouracil, and gemcitabine in advanced pancreatic cancer: a randomised controlled multicentre phase III trial, Lancet Oncol, 6 (2005) 369 - 376. [104] H. Riess, A. Helm, M. Niedergethmann, I. Schmidt-Wolf, M. Moik, C. Hammer, K. Zippel, K. Weigang-Kohler, M. Stauch, H. Oettle, A randomised, prospective, multicenter, phase III trial of gemcitabine, 5-fluorouracil (5- FU), folinic acid vs. gemcitabine alone in patients with advanced pancreatic cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 23 (2005) 4009. [105] C. Rocha Lima, M. Green, R. Rotche, W. Miller, G. Jeffrey, L. Cisar, A. Morganti, N. Orlando, G. Gruia, L. Miller, Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 22 (2004) 3776 - 3783. [106] W. Scheithauer, B. Schull, H. Ulrich-Pur, K. Schmid, M. Raderer, K. Haider, W. Kwasny, D. Depisch, B. Schneeweiss, F. Lang, G. Kornek, Biweekly high- dose gemcitabine alone or in combination with capecitabine in patients with metastatic pancreatic adenocarcinoma: a randomized phase II trial, Ann Oncol, 14 (2003) 97 - 104. [107] G. Stathopoulos, K. Syrigos, G. Aravantinos, A. Polyzos, P. Papakotoulas, G. Fountzilas, A. Potamianou, N. Ziras, J. Boukovinas, J. Varthalitis, N. Androulakis, A. Kotsakis, G. Samonis, V. Georgoulias, A multicenter phase III trial comparing irinotecan-gemcitabine (IG) with gemcitabine (G) monotherapy as first-line treatment in patients with locally advanced or metastatic pancreatic cancer, Br J Cancer, 95 (2006) 587 - 592. [108] H. Ueno, T. Kosuge, Y. Matsuyama, J. Yamamoto, J. Yamamoto, A. Nakao, S. Egawa, R. Doi, M. Monden, T. Hatori, M. Tanaka, M. Shimada, K. Kanemitsu, A randomised phase III trial comparing gemcitabine with surgery-only in patients with

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

resected pancreatic cancer: Japanese Study Group of Adjuvant Therapy for Pancreatic Cancer, Br J Cancer, 101 (2009) 908 - 915. [109] E. Van Cutsem, H. van de Velde, P. Karasek, W. Vervenne, A. Szawlowski, P. Schoffski, S. Post, C. Verslype, H. Neumann, H. Safran, Y. Humblet, J. Perez Ruixo, Y. Ma, D. Von Hoff, Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer, Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 22 (2004) 1430 - 1438. [110] T. Conroy, F. Desseigne, M. Ychou, O. Bouche, R. Guimbaud, Y. Becouarn, A. Adenis, J.L. Raoul, S. Gourgou-Bourgade, C. de la Fouchardiere, J. Bennouna, J.B. Bachet, F. Khemissa-Akouz, D. Pere-Verge, C. Delbaldo, E. Assenat, B. Chauffert, P. Michel, C. Montoto-Grillot, M. Ducreux, U. Groupe Tumeurs Digestives of, P. Intergroup, FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer, The New England journal of medicine, 364 (2011) 1817-1825. [111] D. Papadatos-Pastos, K. Thillai, R. Rabbie, P. Ross, D. Sarker, FOLFIRINOX - a new paradigm in the treatment of pancreatic cancer, Expert review of anticancer therapy, 14 (2014) 1115-1125. [112] D.D. Von Hoff, T. Ervin, F.P. Arena, E.G. Chiorean, J. Infante, M. Moore, T. Seay, S.A. Tjulandin, W.W. Ma, M.N. Saleh, M. Harris, M. Reni, S. Dowden, D. Laheru, N. Bahary, R.K. Ramanathan, J. Tabernero, M. Hidalgo, D. Goldstein, E. Van Cutsem, X. Wei, J. Iglesias, M.F. Renschler, Increased survival in pancreatic cancer with nabpaclitaxel plus gemcitabine, The New England journal of medicine, 369 (2013) 16911703. [113] E. Yu, M.W. Saif, K. Huber, Phase 1 trials in pancreatic cancer, JOP : Journal of the pancreas, 15 (2014) 326-328. [114] S. Ramos, Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention, J Nutr Biochem, 18 (2007) 427-442. [115] R.A. Goldbohm, M.G. Hertog, H.A. Brants, G. van Poppel, P.A. van den Brandt, Consumption of black tea and cancer risk: a prospective cohort study, J Natl Cancer Inst, 88 (1996) 93-100. [116] Y. Tsubono, Y. Nishino, S. Komatsu, C.C. Hsieh, S. Kanemura, I. Tsuji, H. Nakatsuka, A. Fukao, H. Satoh, S. Hisamichi, Green tea and the risk of gastric cancer in Japan, The New England journal of medicine, 344 (2001) 632-636. [117] R. Garcia, C.A. Gonzalez, A. Agudo, E. Riboli, High intake of specific carotenoids and flavonoids does not reduce the risk of bladder cancer, Nutr Cancer, 35 (1999) 212214. [118] I.C. Arts, P.C. Hollman, H.B. Bueno De Mesquita, E.J. Feskens, D. Kromhout, Dietary catechins and epithelial cancer incidence: the Zutphen elderly study, Int J Cancer, 92 (2001) 298-302. [119] R. Garcia-Closas, A. Agudo, C.A. Gonzalez, E. Riboli, Intake of specific carotenoids and flavonoids and the risk of lung cancer in women in Barcelona, Spain, Nutr Cancer, 32 (1998) 154-158. [120] E. De Stefani, H. Deneo-Pellegrini, M. Mendilaharsu, A. Ronco, Diet and risk of cancer of the upper aerodigestive tract--I. Foods, Oral Oncol, 35 (1999) 17-21. [121] K. Imai, K. Suga, K. Nakachi, Cancer-preventive effects of drinking green tea among a Japanese population, Prev Med, 26 (1997) 769-775.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[122] T.J. Key, G.B. Sharp, P.N. Appleby, V. Beral, M.T. Goodman, M. Soda, K. Mabuchi, Soya foods and breast cancer risk: a prospective study in Hiroshima and Nagasaki, Japan, Br J Cancer, 81 (1999) 1248-1256. [123] L.E. Donnelly, R. Newton, G.E. Kennedy, P.S. Fenwick, R.H. Leung, K. Ito, R.E. Russell, P.J. Barnes, Anti-inflammatory effects of resveratrol in lung epithelial cells: molecular mechanisms, Am J Physiol Lung Cell Mol Physiol, 287 (2004) L774-783. [124] B.B. Aggarwal, A. Bhardwaj, R.S. Aggarwal, N.P. Seeram, S. Shishodia, Y. Takada, Role of resveratrol in prevention and therapy of cancer: preclinical and clinical studies, Anticancer research, 24 (2004) 2783-2840. [125] L.M. Hung, J.K. Chen, S.S. Huang, R.S. Lee, M.J. Su, Cardioprotective effect of resveratrol, a natural antioxidant derived from grapes, Cardiovasc Res, 47 (2000) 549555. [126] Z.J. Sun, C.E. Pan, H.S. Liu, G.J. Wang, Anti-hepatoma activity of resveratrol in vitro, World journal of gastroenterology : WJG, 8 (2002) 79-81. [127] G.A. Potter, L.H. Patterson, E. Wanogho, P.J. Perry, P.C. Butler, T. Ijaz, K.C. Ruparelia, J.H. Lamb, P.B. Farmer, L.A. Stanley, M.D. Burke, The cancer preventative agent resveratrol is converted to the anticancer agent piceatannol by the cytochrome P450 enzyme CYP1B1, Br J Cancer, 86 (2002) 774-778. [128] B. Piver, M. Fer, X. Vitrac, J.M. Merillon, Y. Dreano, F. Berthou, D. Lucas, Involvement of cytochrome P450 1A2 in the biotransformation of trans-resveratrol in human liver microsomes, Biochem Pharmacol, 68 (2004) 773-782. [129] J. Lu, C.H. Ho, G. Ghai, K.Y. Chen, Resveratrol analog, 3,4,5,4'tetrahydroxystilbene, differentially induces pro-apoptotic p53/Bax gene expression and inhibits the growth of transformed cells but not their normal counterparts, Carcinogenesis, 22 (2001) 321-328. [130] B. Piver, F. Berthou, Y. Dreano, D. Lucas, Differential inhibition of human cytochrome P450 enzymes by epsilon-viniferin, the dimer of resveratrol: comparison with resveratrol and polyphenols from alcoholized beverages, Life Sci, 73 (2003) 11991213. [131] B. Piver, F. Berthou, Y. Dreano, D. Lucas, Inhibition of CYP3A, CYP1A and CYP2E1 activities by resveratrol and other non volatile red wine components, Toxicol Lett, 125 (2001) 83-91. [132] M.T. Lin, M.L. Yen, C.Y. Lin, M.L. Kuo, Inhibition of vascular endothelial growth factor-induced angiogenesis by resveratrol through interruption of Src-dependent vascular endothelial cadherin tyrosine phosphorylation, Mol Pharmacol, 64 (2003) 10291036. [133] G. Upadhyay, A.K. Singh, A. Kumar, O. Prakash, M.P. Singh, Resveratrol modulates pyrogallol-induced changes in hepatic toxicity markers, xenobiotic metabolizing enzymes and oxidative stress, Eur J Pharmacol, 596 (2008) 146-152. [134] C. Yu, Y.G. Shin, J.W. Kosmeder, J.M. Pezzuto, R.B. van Breemen, Liquid chromatography/tandem mass spectrometric determination of inhibition of human cytochrome P450 isozymes by resveratrol and resveratrol-3-sulfate, Rapid Commun Mass Spectrom, 17 (2003) 307-313. [135] H.P. Ciolino, P.J. Daschner, G.C. Yeh, Resveratrol inhibits transcription of CYP1A1 in vitro by preventing activation of the aryl hydrocarbon receptor, Cancer research, 58 (1998) 5707-5712.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[136] T.K. Chang, J. Chen, W.B. Lee, Differential inhibition and inactivation of human CYP1 enzymes by trans-resveratrol: evidence for mechanism-based inactivation of CYP1A2, J Pharmacol Exp Ther, 299 (2001) 874-882. [137] H. Szaefer, M. Cichocki, D. Brauze, W. Baer-Dubowska, Alteration in phase I and II enzyme activities and polycyclic aromatic hydrocarbons-DNA adduct formation by plant phenolics in mouse epidermis, Nutr Cancer, 48 (2004) 70-77. [138] P. Signorelli, R. Ghidoni, Resveratrol as an anticancer nutrient: molecular basis, open questions and promises, J Nutr Biochem, 16 (2005) 449-466. [139] V. Roman, C. Billard, C. Kern, H. Ferry-Dumazet, J.C. Izard, R. Mohammad, D.M. Mossalayi, J.P. Kolb, Analysis of resveratrol-induced apoptosis in human B-cell chronic leukaemia, Br J Haematol, 117 (2002) 842-851. [140] M. Jang, L. Cai, G.O. Udeani, K.V. Slowing, C.F. Thomas, C.W. Beecher, H.H. Fong, N.R. Farnsworth, A.D. Kinghorn, R.G. Mehta, R.C. Moon, J.M. Pezzuto, Cancer chemopreventive activity of resveratrol, a natural product derived from grapes, Science, 275 (1997) 218-220. [141] O.P. Mgbonyebi, J. Russo, I.H. Russo, Antiproliferative effect of synthetic resveratrol on human breast epithelial cells, International journal of oncology, 12 (1998) 865-869. [142] S.H. Mitchell, W. Zhu, C.Y. Young, Resveratrol inhibits the expression and function of the androgen receptor in LNCaP prostate cancer cells, Cancer research, 59 (1999) 5892-5895. [143] Y. Schneider, F. Vincent, B. Duranton, L. Badolo, F. Gosse, C. Bergmann, N. Seiler, F. Raul, Anti-proliferative effect of resveratrol, a natural component of grapes and wine, on human colonic cancer cells, Cancer letters, 158 (2000) 85-91. [144] J.H. Zhou, H.Y. Cheng, Z.Q. Yu, D.W. He, Z. Pan, D.T. Yang, Resveratrol induces apoptosis in pancreatic cancer cells, Chin Med J (Engl), 124 (2011) 1695-1699. [145] X. Cui, Y. Jin, A.B. Hofseth, E. Pena, J. Habiger, A. Chumanevich, D. Poudyal, M. Nagarkatti, P.S. Nagarkatti, U.P. Singh, L.J. Hofseth, Resveratrol suppresses colitis and colon cancer associated with colitis, Cancer Prev Res (Phila), 3 (2010) 549-559. [146] N. Oi, C.H. Jeong, J. Nadas, Y.Y. Cho, A. Pugliese, A.M. Bode, Z. Dong, Resveratrol, a red wine polyphenol, suppresses pancreatic cancer by inhibiting leukotriene Ahydrolase, Cancer research, 70 (2010) 9755-9764. [147] X. Chen, S. Wang, N. Wu, C.S. Yang, Leukotriene A4 hydrolase as a target for cancer prevention and therapy, Current cancer drug targets, 4 (2004) 267-283. [148] A. Shehzad, F. Wahid, Y.S. Lee, Curcumin in cancer chemoprevention: molecular targets, pharmacokinetics, bioavailability, and clinical trials, Arch Pharm (Weinheim), 343 (2010) 489-499. [149] J. Veeraraghavan, M. Natarajan, P. Lagisetty, V. Awasthi, T.S. Herman, N. Aravindan, Impact of Curcumin, Raspberry Extract, and Neem Leaf Extract on Rel Protein-Regulated Cell Death/Radiosensitization in Pancreatic Cancer Cells, Pancreas, (2011). [150] S. Aggarwal, M.W. Ndinguri, R. Solipuram, N. Wakamatsu, R.P. Hammer, D. Ingram, W. Hansel, [DLys(6) ]-luteinizing hormone releasing hormone-curcumin conjugate inhibits pancreatic cancer cell growth in vitro and in vivo, Int J Cancer, (2011). [151] D. Subramaniam, S. Ramalingam, D.C. Linehan, B.K. Dieckgraefe, R.G. Postier, C.W. Houchen, R.A. Jensen, S. Anant, RNA binding protein CUGBP2/CELF2 mediates

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

curcumin-induced mitotic catastrophe of pancreatic cancer cells, PLoS One, 6 (2011) e16958. [152] W. Glienke, L. Maute, J. Wicht, L. Bergmann, Wilms' tumour gene 1 (WT1) as a target in curcumin treatment of pancreatic cancer cells, Eur J Cancer, 45 (2009) 874-880. [153] W. Glienke, L. Maute, J. Wicht, L. Bergmann, Curcumin inhibits constitutive STAT3 phosphorylation in human pancreatic cancer cell lines and downregulation of survivin/BIRC5 gene expression, Cancer Invest, 28 (2010) 166-171. [154] G. Bar-Sela, R. Epelbaum, M. Schaffer, Curcumin as an anti-cancer agent: review of the gap between basic and clinical applications, Curr Med Chem, 17 (2010) 190-197. [155] R. Epelbaum, M. Schaffer, B. Vizel, V. Badmaev, G. Bar-Sela, Curcumin and gemcitabine in patients with advanced pancreatic cancer, Nutr Cancer, 62 (2010) 11371141. [156] N. Dhillon, B.B. Aggarwal, R.A. Newman, R.A. Wolff, A.B. Kunnumakkara, J.L. Abbruzzese, C.S. Ng, V. Badmaev, R. Kurzrock, Phase II trial of curcumin in patients with advanced pancreatic cancer, Clinical cancer research : an official journal of the American Association for Cancer Research, 14 (2008) 4491-4499. [157] C. Kurbitz, D. Heise, T. Redmer, F. Goumas, A. Arlt, J. Lemke, G. Rimbach, H. Kalthoff, A. Trauzold, Epicatechin gallate and catechin gallate are superior to epigallocatechin gallate in growth suppression and anti-inflammatory activities in pancreatic tumor cells, Cancer Sci, 102 (2011) 728-734. [158] H.A. Vu, Y. Beppu, H.T. Chi, K. Sasaki, H. Yamamoto, P.T. Xinh, T. Tanii, Y. Hara, T. Watanabe, Y. Sato, I. Ohdomari, Green tea epigallocatechin gallate exhibits anticancer effect in human pancreatic carcinoma cells via the inhibition of both focal adhesion kinase and insulin-like growth factor-I receptor, J Biomed Biotechnol, 2010 (2010) 290516. [159] S. Barnes, J. Sfakianos, L. Coward, M. Kirk, Soy isoflavonoids and cancer prevention. Underlying biochemical and pharmacological issues, Adv Exp Med Biol, 401 (1996) 87-100. [160] Y. Li, T. Zhang, Y. Jiang, H.F. Lee, S.J. Schwartz, D. Sun, (-)-Epigallocatechin-3gallate inhibits Hsp90 function by impairing Hsp90 association with cochaperones in pancreatic cancer cell line Mia Paca-2, Mol Pharm, 6 (2009) 1152-1159. [161] A. Basu, S. Haldar, Combinatorial effect of epigallocatechin-3-gallate and TRAIL on pancreatic cancer cell death, International journal of oncology, 34 (2009) 281-286. [162] S. Shankar, S. Ganapathy, S.R. Hingorani, R.K. Srivastava, EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer, Front Biosci, 13 (2008) 440452. [163] P.B. Kaufman, J.A. Duke, H. Brielmann, J. Boik, J.E. Hoyt, A comparative survey of leguminous plants as sources of the isoflavones, genistein and daidzein: implications for human nutrition and health, J Altern Complement Med, 3 (1997) 7-12. [164] C.V. Rao, K. Tokumo, J. Rigotty, E. Zang, G. Kelloff, B.S. Reddy, Chemoprevention of colon carcinogenesis by dietary administration of piroxicam, alphadifluoromethylornithine, 16 alpha-fluoro-5-androsten-17-one, and ellagic acid individually and in combination, Cancer research, 51 (1991) 4528-4534. [165] R.C. Alves, I.M. Almeida, S. Casal, M.B. Oliveira, Isoflavones in coffee: influence of species, roast degree, and brewing method, J Agric Food Chem, 58 (2010) 3002-3007.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[166] T. Akiyama, J. Ishida, S. Nakagawa, H. Ogawara, S. Watanabe, N. Itoh, M. Shibuya, Y. Fukami, Genistein, a specific inhibitor of tyrosine-specific protein kinases, J Biol Chem, 262 (1987) 5592-5595. [167] Z.C. Dang, V. Audinot, S.E. Papapoulos, J.A. Boutin, C.W. Lowik, Peroxisome proliferator-activated receptor gamma (PPARgamma ) as a molecular target for the soy phytoestrogen genistein, J Biol Chem, 278 (2003) 962-967. [168] G. Gossner, M. Choi, L. Tan, S. Fogoros, K.A. Griffith, M. Kuenker, J.R. Liu, Genistein-induced apoptosis and autophagocytosis in ovarian cancer cells, Gynecol Oncol, 105 (2007) 23-30. [169] G.G. Kuiper, J.G. Lemmen, B. Carlsson, J.C. Corton, S.H. Safe, P.T. van der Saag, B. van der Burg, J.A. Gustafsson, Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta, Endocrinology, 139 (1998) 4252-4263. [170] M. Lopez-Lazaro, E. Willmore, C.A. Austin, Cells lacking DNA topoisomerase II beta are resistant to genistein, J Nat Prod, 70 (2007) 763-767. [171] G.E. Mann, B. Bonacasa, T. Ishii, R.C. Siow, Targeting the redox sensitive Nrf2Keap1 defense pathway in cardiovascular disease: protection afforded by dietary isoflavones, Curr Opin Pharmacol, 9 (2009) 139-145. [172] J. Markovits, C. Linassier, P. Fosse, J. Couprie, J. Pierre, A. Jacquemin-Sablon, J.M. Saucier, J.B. Le Pecq, A.K. Larsen, Inhibitory effects of the tyrosine kinase inhibitor genistein on mammalian DNA topoisomerase II, Cancer Res, 49 (1989) 5111-5117. [173] Y. Nakamura, S. Yogosawa, Y. Izutani, H. Watanabe, E. Otsuji, T. Sakai, A combination of indol-3-carbinol and genistein synergistically induces apoptosis in human colon cancer HT-29 cells by inhibiting Akt phosphorylation and progression of autophagy, Mol Cancer, 8 (2009) 100. [174] J.C. Vera, A.M. Reyes, J.G. Carcamo, F.V. Velasquez, C.I. Rivas, R.H. Zhang, P. Strobel, R. Iribarren, H.I. Scher, J.C. Slebe, et al., Genistein is a natural inhibitor of hexose and dehydroascorbic acid transport through the glucose transporter, GLUT1, J Biol Chem, 271 (1996) 8719-8724. [175] A. Constantinou, K. Kiguchi, E. Huberman, Induction of differentiation and DNA strand breakage in human HL-60 and K-562 leukemia cells by genistein, Cancer Res, 50 (1990) 2618-2624. [176] T. Fotsis, M. Pepper, H. Adlercreutz, G. Fleischmann, T. Hase, R. Montesano, L. Schweigerer, Genistein, a dietary-derived inhibitor of in vitro angiogenesis, Proc Natl Acad Sci U S A, 90 (1993) 2690-2694. [177] W. Gorczyca, J. Gong, B. Ardelt, F. Traganos, Z. Darzynkiewicz, The cell cycle related differences in susceptibility of HL-60 cells to apoptosis induced by various antitumor agents, Cancer Res, 53 (1993) 3186-3192. [178] A. Kuriu, H. Ikeda, Y. Kanakura, J.D. Griffin, B. Druker, H. Yagura, H. Kitayama, J. Ishikawa, T. Nishiura, Y. Kanayama, et al., Proliferation of human myeloid leukemia cell line associated with the tyrosine-phosphorylation and activation of the protooncogene c-kit product, Blood, 78 (1991) 2834-2840. [179] B.Q. Li, M. Subleski, D. Shalloway, H.F. Kung, T. Kamata, Mitogenic activation of the Ras guanine nucleotide exchange factor in NIH 3T3 cells involves protein tyrosine phosphorylation, Proc Natl Acad Sci U S A, 90 (1993) 8504-8508.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[180] C. Linassier, M. Pierre, J.B. Le Pecq, J. Pierre, Mechanisms of action in NIH-3T3 cells of genistein, an inhibitor of EGF receptor tyrosine kinase activity, Biochem Pharmacol, 39 (1990) 187-193. [181] D. Linnekin, L.S. Park, W.L. Farrar, Dissociation of human cytokine receptor expression and signal transduction, Blood, 80 (1992) 1896-1904. [182] Y. Matsukawa, N. Marui, T. Sakai, Y. Satomi, M. Yoshida, K. Matsumoto, H. Nishino, A. Aoike, Genistein arrests cell cycle progression at G2-M, Cancer Res, 53 (1993) 1328-1331. [183] A. Okura, H. Arakawa, H. Oka, T. Yoshinari, Y. Monden, Effect of genistein on topoisomerase activity and on the growth of [Val 12]Ha-ras-transformed NIH 3T3 cells, Biochem Biophys Res Commun, 157 (1988) 183-189. [184] F. Traganos, B. Ardelt, N. Halko, S. Bruno, Z. Darzynkiewicz, Effects of genistein on the growth and cell cycle progression of normal human lymphocytes and human leukemic MOLT-4 and HL-60 cells, Cancer Res, 52 (1992) 6200-6208. [185] B. Bao, Z. Wang, S. Ali, D. Kong, S. Banerjee, A. Ahmad, Y. Li, A.S. Azmi, L. Miele, F.H. Sarkar, Over-expression of FoxM1 leads to epithelial-mesenchymal transition and cancer stem cell phenotype in pancreatic cancer cells, J Cell Biochem, (2011). [186] Z. Wang, A. Ahmad, S. Banerjee, A. Azmi, D. Kong, Y. Li, F.H. Sarkar, FoxM1 is a novel target of a natural agent in pancreatic cancer, Pharm Res, 27 (2010) 1159-1168. [187] J.W. Fahey, Y. Zhang, P. Talalay, Broccoli sprouts: an exceptionally rich source of inducers of enzymes that protect against chemical carcinogens, Proceedings of the National Academy of Sciences of the United States of America, 94 (1997) 10367-10372. [188] Y. Zhang, T.W. Kensler, C.G. Cho, G.H. Posner, P. Talalay, Anticarcinogenic activities of sulforaphane and structurally related synthetic norbornyl isothiocyanates, Proceedings of the National Academy of Sciences of the United States of America, 91 (1994) 3147-3150. [189] F.L. Chung, C.C. Conaway, C.V. Rao, B.S. Reddy, Chemoprevention of colonic aberrant crypt foci in Fischer rats by sulforaphane and phenethyl isothiocyanate, Carcinogenesis, 21 (2000) 2287-2291. [190] G. Shen, T.O. Khor, R. Hu, S. Yu, S. Nair, C.T. Ho, B.S. Reddy, M.T. Huang, H.L. Newmark, A.N. Kong, Chemoprevention of familial adenomatous polyposis by natural dietary compounds sulforaphane and dibenzoylmethane alone and in combination in ApcMin/+ mouse, Cancer research, 67 (2007) 9937-9944. [191] D. Chaudhuri, S. Orsulic, B.T. Ashok, Antiproliferative activity of sulforaphane in Akt-overexpressing ovarian cancer cells, Molecular cancer therapeutics, 6 (2007) 334345. [192] S.Y. Park, G.Y. Kim, S.J. Bae, Y.H. Yoo, Y.H. Choi, Induction of apoptosis by isothiocyanate sulforaphane in human cervical carcinoma HeLa and hepatocarcinoma HepG2 cells through activation of caspase-3, Oncology reports, 18 (2007) 181-187. [193] Y. Li, T. Zhang, H. Korkaya, S. Liu, H.F. Lee, B. Newman, Y. Yu, S.G. Clouthier, S.J. Schwartz, M.S. Wicha, D. Sun, Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells, Clinical cancer research : an official journal of the American Association for Cancer Research, 16 (2010) 2580-2590.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[194] Y. Li, T. Zhang, S.J. Schwartz, D. Sun, Sulforaphane potentiates the efficacy of 17allylamino 17-demethoxygeldanamycin against pancreatic cancer through enhanced abrogation of Hsp90 chaperone function, Nutr Cancer, 63 (2011) 1151-1159. [195] R.K. Srivastava, S.N. Tang, W. Zhu, D. Meeker, S. Shankar, Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells, Frontiers in bioscience, 3 (2011) 515-528. [196] G. Kallifatidis, V. Rausch, B. Baumann, A. Apel, B.M. Beckermann, A. Groth, J. Mattern, Z. Li, A. Kolb, G. Moldenhauer, P. Altevogt, T. Wirth, J. Werner, P. Schemmer, M.W. Buchler, A.V. Salnikov, I. Herr, Sulforaphane targets pancreatic tumour-initiating cells by NF-kappaB-induced antiapoptotic signalling, Gut, 58 (2009) 949-963. [197] Y. Shukla, N. Kalra, Cancer chemoprevention with garlic and its constituents, Cancer letters, 247 (2007) 167-181. [198] C.V. Rao, Benzyl isothiocyanate: double trouble for breast cancer cells, Cancer Prev Res (Phila), 6 (2013) 760-763. [199] S.S. Hecht, Chemoprevention of cancer by isothiocyanates, modifiers of carcinogen metabolism, J Nutr, 129 (1999) 768S-774S. [200] S.S. Hecht, Inhibition of carcinogenesis by isothiocyanates, Drug Metab Rev, 32 (2000) 395-411. [201] L.W. Wattenberg, Inhibition of carcinogenic effects of polycyclic hydrocarbons by benzyl isothiocyanate and related compounds, J Natl Cancer Inst, 58 (1977) 395-398. [202] L.W. Wattenberg, Inhibition of carcinogen-induced neoplasia by sodium cyanate, tert-butyl isocyanate, and benzyl isothiocyanate administered subsequent to carcinogen exposure, Cancer Res, 41 (1981) 2991-2994. [203] Y. Zhang, P. Talalay, C.G. Cho, G.H. Posner, A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure, Proc Natl Acad Sci U S A, 89 (1992) 2399-2403. [204] S. Batra, R.P. Sahu, P.K. Kandala, S.K. Srivastava, Benzyl isothiocyanatemediated inhibition of histone deacetylase leads to NF-kappaB turnoff in human pancreatic carcinoma cells, Mol Cancer Ther, 9 (2010) 1596-1608. [205] R.P. Sahu, S.K. Srivastava, The role of STAT-3 in the induction of apoptosis in pancreatic cancer cells by benzyl isothiocyanate, J Natl Cancer Inst, 101 (2009) 176-193. [206] R.P. Sahu, R. Zhang, S. Batra, Y. Shi, S.K. Srivastava, Benzyl isothiocyanatemediated generation of reactive oxygen species causes cell cycle arrest and induces apoptosis via activation of MAPK in human pancreatic cancer cells, Carcinogenesis, 30 (2009) 1744-1753. [207] R. Zhang, S. Loganathan, I. Humphreys, S.K. Srivastava, Benzyl isothiocyanateinduced DNA damage causes G2/M cell cycle arrest and apoptosis in human pancreatic cancer cells, J Nutr, 136 (2006) 2728-2734. [208] S.R. Boreddy, R.P. Sahu, S.K. Srivastava, Benzyl isothiocyanate suppresses pancreatic tumor angiogenesis and invasion by inhibiting HIF-alpha/VEGF/RhoGTPases: pivotal role of STAT-3, PLoS One, 6 (2011) e25799. [209] S.R. Boreddy, K.C. Pramanik, S.K. Srivastava, Pancreatic tumor suppression by benzyl isothiocyanate is associated with inhibition of PI3K/AKT/FOXO pathway, Clin Cancer Res, 17 (2011) 1784-1795. [210] L. Raj, T. Ide, A.U. Gurkar, M. Foley, M. Schenone, X. Li, N.J. Tolliday, T.R. Golub, S.A. Carr, A.F. Shamji, A.M. Stern, A. Mandinova, S.L. Schreiber, S.W. Lee,

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

Selective killing of cancer cells by a small molecule targeting the stress response to ROS, Nature, 475 (2011) 231-234. [211] H. Randhawa, K. Kibble, H. Zeng, M.P. Moyer, K.M. Reindl, Activation of ERK signaling and induction of colon cancer cell death by piperlongumine, Toxicol In Vitro, 27 (2013) 1626-1633. [212] H. Dhillon, S. Chikara, K.M. Reindl, Piperlongumine induces pancreatic cancer cell death by enhancing reactive oxygen species and DNA damage, Toxicol Rep, 1 (2014) 309-318. [213] Y. Qin, Z. Ma, X. Dang, W. Li, Q. Ma, Effect of resveratrol on proliferation and apoptosis of human pancreatic cancer MIA PaCa-2 cells may involve inhibition of the Hedgehog signaling pathway, Molecular medicine reports, 10 (2014) 2563-2567. [214] A. Kumar, J. Hu, H.A. LaVoie, K.B. Walsh, D.J. DiPette, U.S. Singh, Conformational changes and translocation of tissue-transglutaminase to the plasma membranes: role in cancer cell migration, BMC cancer, 14 (2014) 256. [215] L. Yang, L. Yang, W. Tian, J. Li, J. Liu, M. Zhu, Y. Zhang, Y. Yang, F. Liu, Q. Zhang, Q. Liu, Y. Shen, Z. Qi, Resveratrol plays dual roles in pancreatic cancer cells, Journal of cancer research and clinical oncology, 140 (2014) 749-755. [216] W. Li, J. Ma, Q. Ma, B. Li, L. Han, J. Liu, Q. Xu, W. Duan, S. Yu, F. Wang, E. Wu, Resveratrol inhibits the epithelial-mesenchymal transition of pancreatic cancer cells via suppression of the PI-3K/Akt/NF-kappaB pathway, Curr Med Chem, 20 (2013) 41854194. [217] P. Liu, H. Liang, Q. Xia, P. Li, H. Kong, P. Lei, S. Wang, Z. Tu, Resveratrol induces apoptosis of pancreatic cancers cells by inhibiting miR-21 regulation of BCL-2 expression, Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico, 15 (2013) 741-746. [218] W. Mo, X. Xu, L. Xu, F. Wang, A. Ke, X. Wang, C. Guo, Resveratrol inhibits proliferation and induces apoptosis through the hedgehog signaling pathway in pancreatic cancer cell, Pancreatology : official journal of the International Association of Pancreatology, 11 (2011) 601-609. [219] S.K. Roy, Q. Chen, J. Fu, S. Shankar, R.K. Srivastava, Resveratrol inhibits growth of orthotopic pancreatic tumors through activation of FOXO transcription factors, PLoS One, 6 (2011) e25166. [220] S. Bimonte, A. Barbieri, G. Palma, A. Luciano, D. Rea, C. Arra, Curcumin inhibits tumor growth and angiogenesis in an orthotopic mouse model of human pancreatic cancer, BioMed research international, 2013 (2013) 810423. [221] S. Sarkar, H. Dubaybo, S. Ali, P. Goncalves, S.L. Kollepara, S. Sethi, P.A. Philip, Y. Li, Down-regulation of miR-221 inhibits proliferation of pancreatic cancer cells through up-regulation of PTEN, p27(kip1), p57(kip2), and PUMA, American journal of cancer research, 3 (2013) 465-477. [222] A.P. Ranjan, A. Mukerjee, L. Helson, R. Gupta, J.K. Vishwanatha, Efficacy of liposomal curcumin in a human pancreatic tumor xenograft model: inhibition of tumor growth and angiogenesis, Anticancer research, 33 (2013) 3603-3609. [223] M. Youns, G.M. Fathy, Upregulation of extrinsic apoptotic pathway in curcuminmediated antiproliferative effect on human pancreatic carcinogenesis, J Cell Biochem, 114 (2013) 2654-2665.

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

[224] X.D. Sun, X.E. Liu, D.S. Huang, Curcumin reverses the epithelial-mesenchymal transition of pancreatic cancer cells by inhibiting the Hedgehog signaling pathway, Oncology reports, 29 (2013) 2401-2407. [225] B. Bao, S. Ali, S. Banerjee, Z. Wang, F. Logna, A.S. Azmi, D. Kong, A. Ahmad, Y. Li, S. Padhye, F.H. Sarkar, Curcumin analogue CDF inhibits pancreatic tumor growth by switching on suppressor microRNAs and attenuating EZH2 expression, Cancer research, 72 (2012) 335-345. [226] S. Liu, X.J. Wang, Y. Liu, Y.F. Cui, PI3K/AKT/mTOR signaling is involved in (-)epigallocatechin-3-gallate-induced apoptosis of human pancreatic carcinoma cells, The American journal of Chinese medicine, 41 (2013) 629-642. [227] S.O. Kim, M.R. Kim, (-)-Epigallocatechin 3-gallate inhibits invasion by inducing the expression of Raf kinase inhibitor protein in AsPC1 human pancreatic adenocarcinoma cells through the modulation of histone deacetylase activity, International journal of oncology, 42 (2013) 349-358. [228] C. Hardtner, G. Multhoff, W. Falk, J. Radons, (-)-Epigallocatechin-3-gallate, a green tea-derived catechin, synergizes with celecoxib to inhibit IL-1-induced tumorigenic mediators by human pancreatic adenocarcinoma cells Colo357, European journal of pharmacology, 684 (2012) 36-43. [229] Z. Zhu, Y. Wang, Z. Liu, F. Wang, Q. Zhao, Inhibitory effects of epigallocatechin3-gallate on cell proliferation and the expression of HIF-1alpha and P-gp in the human pancreatic carcinoma cell line PANC-1, Oncology reports, 27 (2012) 1567-1572. [230] S.N. Tang, J. Fu, S. Shankar, R.K. Srivastava, EGCG enhances the therapeutic potential of gemcitabine and CP690550 by inhibiting STAT3 signaling pathway in human pancreatic cancer, PLoS One, 7 (2012) e31067. [231] S.N. Tang, J. Fu, D. Nall, M. Rodova, S. Shankar, R.K. Srivastava, Inhibition of sonic hedgehog pathway and pluripotency maintaining factors regulate human pancreatic cancer stem cell characteristics, Int J Cancer, 131 (2012) 30-40. [232] T. Forster, V. Rausch, Y. Zhang, O. Isayev, K. Heilmann, F. Schoensiegel, L. Liu, M. Nessling, K. Richter, S. Labsch, C.C. Nwaeburu, J. Mattern, J. Gladkich, N. Giese, J. Werner, P. Schemmer, W. Gross, M.M. Gebhard, C. Gerhauser, M. Schaefer, I. Herr, Sulforaphane counteracts aggressiveness of pancreatic cancer driven by dysregulated Cx43-mediated gap junctional intercellular communication, Oncotarget, 5 (2014) 16211634. [233] A. Thakkar, D. Sutaria, B.K. Grandhi, J. Wang, S. Prabhu, The molecular mechanism of action of aspirin, curcumin and sulforaphane combinations in the chemoprevention of pancreatic cancer, Oncology reports, 29 (2013) 1671-1677. [234] Y. Li, G.E. Karagoz, Y.H. Seo, T. Zhang, Y. Jiang, Y. Yu, A.M. Duarte, S.J. Schwartz, R. Boelens, K. Carroll, S.G. Rudiger, D. Sun, Sulforaphane inhibits pancreatic cancer through disrupting Hsp90-p50(Cdc37) complex and direct interactions with amino acids residues of Hsp90, J Nutr Biochem, 23 (2012) 1617-1626. [235] G. Kallifatidis, S. Labsch, V. Rausch, J. Mattern, J. Gladkich, G. Moldenhauer, M.W. Buchler, A.V. Salnikov, I. Herr, Sulforaphane increases drug-mediated cytotoxicity toward cancer stem-like cells of pancreas and prostate, Molecular therapy : the journal of the American Society of Gene Therapy, 19 (2011) 188-195.

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

TE

D

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT Disclosure of Potential Conflicts of Interest

AC CE P

TE

D

MA

NU

SC

RI

PT

The authors have declared that no Conflicts of Interest exist.

ACCEPTED MANUSCRIPT Highlights 

Pancreatic cancer (PC) is a complex hereditary disease with high mortality

PT

rate.

PC exhibits resistance against chemo- and radiotherapy.



PC stem cells contribute greatly to its resistance against chemo- and

SC

RI



Herbal products target multiple pathways involved in carcinogenesis

MA



NU

radiotherapy.

simultaneously.

TE

cure.

D

Solution involving herbals may be a smart thought for better results in PC

AC CE P



Recent advances in pancreatic cancer: biology, treatment, and prevention.

Pancreatic cancer (PC) is the fourth leading cause of cancer-related death in United States. Efforts have been made towards the development of the via...
681KB Sizes 7 Downloads 11 Views