+ All Categories
Home > Documents > BBA Rev on Cancer

BBA Rev on Cancer

Date post: 17-Aug-2015
Category:
Upload: ghanshyam-upadhyay
View: 63 times
Download: 2 times
Share this document with a friend
Popular Tags:
15
Review Recent advances in pancreatic cancer: biology, treatment, and prevention Divya Singh a , Ghanshyam Upadhyay a, , Rakesh K. Srivastava b, , Sharmila Shankar b,c, ⁎⁎ a Department of Biology, City College of New York, 160 Convent Avenue, New York, NY 10031, USA b Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128, USA c Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA abstract article info Article history: Received 30 January 2015 Received in revised form 28 April 2015 Accepted 30 April 2015 Available online 11 May 2015 Keywords: Pancreatic cancer Cancer stem cell Signaling pathways Cancer prevention Natural Product Pancreatic cancer (PC) is the fourth leading cause of cancer-related death in United States. Efforts have been made towards the development of the viable solution for its treatment with constrained accomplishment because of its complex biology. It is well established that pancreatic cancer stem cells (CSCs), albeit present in a little count, contribute incredibly to PC initiation, progression, and metastasis. Customary chemo and radiotherapeutic alternatives, however, expands general survival, the related side effects are the signicant 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 enormously. Presently the focus is to target CSCs. The herbal products have gained much consideration recently as they, usually, sensitize CSCs to chemotherapy and target molecular signaling involved in various tumors including PC. Some planned studies have 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. © 2015 Elsevier B.V. All rights reserved. Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 2. Biology of pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 3. Signaling pathways in pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16 4. STAT3 pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16 5. Smad/TGF-β pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16 6. Wnt pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 Biochimica et Biophysica Acta 1856 (2015) 1327 Abbreviations: Oct4,octamer-bindingtranscription factor 4;ABCG2,ATP-bindingcassette sub-family G member 2; CXCR4,C-X-C chemokinereceptor type4;FGF, broblastgrowthfactor; Frizzled-9, frizzled class receptor 9; Glut1, glucose transporter 1; Foxa2, forkhead box A2; Sox2, sex determining region Y box 2; Klf4, kruppel like factor 4; c-Myc, v-Myc avian myelocytomatosis viral oncogene homolog; FGF, broblast growth factor; ESA, epithelial-specic antigen; ALDH1, acetaldehyde dehydrogenases 1; ABCB1, ATP-binding cassette sub- family B member 1; MDR1, multidrug resistance protein 1; DCLK1, doublecortin-like kinase 1; Cdkn2a, cyclin-dependent kinase inhibitor 2a; Dpc4 or Smad4, deleted in pancreatic carcinoma, locus 4; STAT3, signal transducer and activator of transcription 3; TNF-α, tumor necrosis factor α; MCP-1, monocyte chemotactic protein-1; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; PDGF, platelet-derived growth factor; G-CSF, granulocyte colony-stimulating factor; 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, hyp- oxiainduciblefactor 1α;MMP,matrixmetalloproteinase,TWIST1,Twist-relatedprotein1;ICAM1,intercellularadhesion molecule1; Bcl-2, B-celllymphoma2, Bcl-xL,B-celllymphomaextra- large,Bad,Bcl-2-associateddeathpromoter;Bak,Bcl-2homologousantagonist/killer;Bax,Bcl-2-associatedXprotein;Mcl-1,inducedmyeloidleukemiacelldifferentiationprotein;Pdx1,pan- creatic 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, forkhead box O3; PI3K, phosphatidylinositol 3-kinase; PARP, peroxisome proliferator-activated receptor; PTEN, phosphatase and tensin homolog; PDGFRα, alpha-type platelet-derived growth factor receptor; IGF2R, insulin-like growth factor 2 receptor; 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, tumor necrosis factor-related apoptosis-inducing ligand; Cdk4, cyclin-dependent kinase 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 receptor; EpCAM, epithelial cell ad- hesion molecule; vWF, von Willebrand factor; PCNA, proliferating cell nuclear antigen; Hsp, heat shock potein; XIAP, X-linked inhibitor of apoptosis protein; IAP, inhibitor of apoptosis protein; Pdk1, phosphoinositide-dependent kinase-1; mTOR, mammalian target of rapamycin; ERK, extracellular-signal-regulated kinases; JNK, c-Jun N-terminal kinases; HDAC, histone deacetylases; p38, P38 mitogen-activated protein kinases; ROS, reactive oxygen species. Corresponding author. ⁎⁎ Correspondence to: S. Shankar, Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128. E-mail addresses: [email protected] (D. Singh), [email protected] (G. Upadhyay), [email protected] (R.K. Srivastava), [email protected] (S. Shankar). http://dx.doi.org/10.1016/j.bbcan.2015.04.003 0304-419X/© 2015 Elsevier B.V. All rights reserved. Contents lists available at ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbacan
Transcript
Page 1: BBA Rev on Cancer

Biochimica et Biophysica Acta 1856 (2015) 13–27

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta

j ourna l homepage: www.e lsev ie r .com/ locate /bbacan

Review

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

Divya Singh a, Ghanshyam Upadhyay a,⁎, Rakesh K. Srivastava b,⁎, Sharmila Shankar b,c,⁎⁎a Department of Biology, City College of New York, 160 Convent Avenue, New York, NY 10031, USAb Kansas City VA Medical Center, 4801 Linwood Boulevard, Kansas City, MO 64128, USAc Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA

Abbreviations:Oct4,octamer-bindingtranscriptionfactFrizzled-9, frizzled class receptor 9; Glut1, glucose tranmyelocytomatosis viral oncogene homolog; FGF, fibroblafamilyBmember1;MDR1,multidrugresistanceprotein1;Dlocus 4; STAT3, signal transducer and activator of transcripgrowth factor receptor; PDGF, platelet-derived growthtransforminggrowth factorbeta; Chk2, checkpoint kinase2oxiainduciblefactor1α;MMP,matrixmetalloproteinase,Tlarge,Bad,Bcl-2-associateddeathpromoter;Bak,Bcl-2homcreatic and duodenal homeobox 1, uPA, urokinase-type pforkhead box O1; FoxO3, forkhead box O3; PI3K, phosphalpha-type platelet-derived growth factor receptor; IGF2forkhead box O1; AFX, forkhead box O4; TP53, tumor prooncogene serine/threonine-protein kinase;Her-2, humanhesionmolecule;vWF,vonWillebrandfactor;PCNA,prolifePdk1, phosphoinositide-dependent kinase-1; mTOR, mdeacetylases; p38, P38mitogen-activated protein kinases;⁎ Corresponding author.⁎⁎ Correspondence to: S. Shankar, Kansas City VA Medic

E-mail addresses: [email protected] (D. Sin(S. Shankar).

http://dx.doi.org/10.1016/j.bbcan.2015.04.0030304-419X/© 2015 Elsevier B.V. All rights reserved.

a b s t r a c t

a r t i c l e i n f o

Article history:Received 30 January 2015Received in revised form 28 April 2015Accepted 30 April 2015Available online 11 May 2015

Keywords:Pancreatic cancerCancer stem cellSignaling pathwaysCancer preventionNatural Product

Pancreatic cancer (PC) is the fourth leading cause of cancer-related death in United States. Efforts have beenmade towards the development of the viable solution for its treatment with constrained accomplishmentbecause of its complex biology. It is well established that pancreatic cancer stem cells (CSCs), albeit present ina little count, contribute incredibly to PC initiation, progression, and metastasis. Customary chemo andradiotherapeutic alternatives, however, expands general survival, the related side effects are the significantconcern. Amid the most recent decade, our insight about molecular and cellular pathways involved in PC androle of CSCs in its progression has increased enormously. Presently the focus is to target CSCs. The herbal productshave gained much consideration recently as they, usually, sensitize CSCs to chemotherapy and target molecularsignaling involved in various tumors including PC. Some planned studies have indicated promising resultsproposing that examinations in this course have a lot to offer for the treatment of PC. Although preclinical studiesuncovered the importance of herbal products in attenuating pancreatic carcinoma, limited studies have beenconducted to evaluate their role in clinics. The present review provides a new insight to recent advances inpancreatic cancer biology, treatment and current status of herbal products in its anticipation.

© 2015 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 142. Biology of pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143. Signaling pathways in pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 164. STAT3 pathway. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 165. Smad/TGF-β pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166. Wnt pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

or4;ABCG2,ATP-bindingcassettesub-familyGmember2;CXCR4,C-X-Cchemokinereceptortype4;FGF,fibroblastgrowthfactor;sporter 1; Foxa2, forkhead box A2; Sox2, sex determining region Y box 2; Klf4, kruppel like factor 4; c-Myc, v-Myc avianst growth factor; ESA, epithelial-specific antigen; ALDH1, acetaldehyde dehydrogenases 1; ABCB1, ATP-binding cassette sub-CLK1,doublecortin-likekinase1;Cdkn2a,cyclin-dependentkinase inhibitor2a;Dpc4orSmad4,deleted inpancreaticcarcinoma,

tion 3; TNF-α, tumor necrosis factorα;MCP-1,monocyte chemotactic protein-1; EGF, epidermal growth factor; EGFR, epidermalfactor; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; TGF-β,; COX-2, cyclooxygenase-2; IGF-1R, insulin-like growth factor-1 receptor; VEGF, vascular endothelial growth factor;HIF1α, hyp-

WIST1,Twist-relatedprotein1;ICAM1,intercellularadhesionmolecule1;Bcl-2,B-celllymphoma2,Bcl-xL,B-celllymphomaextra-ologousantagonist/killer;Bax,Bcl-2-associatedXprotein;Mcl-1, inducedmyeloidleukemiacelldifferentiationprotein;Pdx1,pan-lasminogen activator; uPAR, urokinase-type plasminogen activator receptor; MAPK,mitogen activated protein kinase; FoxO1,atidylinositol 3-kinase; PARP, peroxisome proliferator-activated receptor; PTEN, phosphatase and tensin homolog; PDGFRα,R, insulin-like growth factor 2 receptor; ENG, endoglin, ALK1, activin receptor-like kinase 1; FKHRL1, forkhead box O3a; FKHR,tein p53; TRAIL, tumor necrosis factor-related apoptosis-inducing ligand; Cdk4, cyclin-dependent kinase 4; Raf-1, RAF proto-epidermal growth factor receptor 2; EMT, endothelial tomeseanchymal transition;DR, death receptor; EpCAM, epithelial cell ad-ratingcellnuclearantigen;Hsp,heatshockpotein;XIAP,X-linkedinhibitorofapoptosisprotein;IAP, inhibitorofapoptosisprotein;ammalian target of rapamycin; ERK, extracellular-signal-regulated kinases; JNK, c-Jun N-terminal kinases; HDAC, histoneROS, reactive oxygen species.

al Center, 4801 Linwood Boulevard, Kansas City, MO 64128.gh), [email protected] (G. Upadhyay), [email protected] (R.K. Srivastava), [email protected]

Page 2: BBA Rev on Cancer

14 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

7. Notch pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178. PI3K/Akt pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189. Sonic Hedgehog pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1810. Treatment of pancreatic cancer: chemotherapy and radiotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1811. Clinical trials in pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1812. Herbal products, cancer prevention, and pancreatic cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1913. Resveratrol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2014. Curcumin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2015. Epigallocatechin gallate (EGCG) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2116. Genistein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2117. Sulforaphane . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2218. Garlic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2219. Benzyl isothiocyanate (BITC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2220. Piperlongumine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2321. Conclusion and future perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23Disclosure of potential conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23

1. Introduction

The burden of pancreatic cancer (PC) has continuously increasedworldwide. It is a serious health concern and fourth leading causeof cancer-related death in United States of America [1,2]. PC is describedas a type of gastrointestinal tumor with a poor anticipation and a highlevel of danger and death rate [3]. More than 90% of pancreatic tumorshave inception from the ductal epithelium of pancreas consequentlytermed as pancreatic ductal adenocarcinoma (PDAC). It is disturbingto see that frequency rate of the pancreatic tumor is relentlesslyexpanding in the western world [4]. The danger components for pan-creatic cancer incorporate smoking, obesity and high utilization ofprocessed meat. Age is positively correlated with pancreatic cancerincidences, and the larger part of cases are diagnosed over the ageof 60 [5]. The introductory indications of patients with PDAC areback agony and dyspepsia with additional disturbing manifestationslike the new onset of diabetes, jaundice, unconstrained profoundvein thrombosis and weight reduction. When one begins perceiving,the tumor typically, spreads to the encompassing tissues or distantorgans. For the tumors spotted in the head region of pancreas, thedetermination is actually productive and they are diagnosed rela-tively early because of biliary impediment. Nonetheless, the tumorsin the body and tail of pancreas regularly stay asymptomatic untillate in disease stage. Most of the patients (~80%) are identifiedwith unresectable locally advanced or metastatic stage and themajor cause is the delayed diagnosis and lack of specific bloodor urine biomarkers to identify patients with increased risk of devel-oping pancreatic cancer [6–9]. The routine diagnostics incorporatetransabdominal ultrasound in the introductory assessment of thejaundiced patient alongside computed tomography (CT) scan ormagnet-ic resonance imaging (MRI).

Despite the fact that the survival rate for most cancers has beenincreased lately in a couple of decades, little change is seen in the caseof pancreatic cancer. The usual survival rate for pancreatic cancerpatients is under six months, and just 3% patients survive over 5-years[6–9]. The reason is attributed to various factors including silent naturein early stages, aggressive tumor biology, the low scope of surgicalmanagement, and lack of effective systemic therapies. Although, thecurrent procedures including surgery, chemotherapy, radiation, and im-munosuppressants, have made great advances in diminishing tumorfrequencies and death rates, pancreatic cancer remains a continuingchallenge to the researchers. The treatment strategies at present utilizedare not very encouraging [10]. There are exceptionally poor post-surgery survival rates even when the pancreatic tumor is surgicallyresected. Safety concerns related with these medications/techniquesare likewise a significant issue for their accomplishment in the treatment

of the disease [6–9]. The prevalent chemotherapeutic choices for thecancer treatment prolong the life of pancreatic cancer patientsminimally,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 showsresistance against chemo- and radiotherapies, it is important to findnovel and viable methodologies for the treatment of pancreatic cancer[10].

Although the potential use of herbal components for the protectionagainst various cancers began several decades ago, studies to under-stand the mechanism of their action at biochemical, genomic, and pro-teomic levels started very recently. Many plant products, such astriterpenes, flavonoids or polyphenols, are now established potent che-mopreventive agents [11–15]. The phenolic substances are isolatedfrom the wide range of vascular plants and have the ability to reduceand scavenge free radicals [16,17]. Epidemiological studies haveshown the reduced risk of pancreatic cancer by increased consumptionof fruits and vegetables [18]. In the recent past, a number of preclinicalstudies have demonstrated various degrees of the efficacy of herbalproducts both in vitro and in vivo [18]. Certain dietary agents, for exam-ple, resveratrol and curcumin, have been demonstrated to potentiatethe standard chemotherapy [18]. It has been observed that herbal prod-ucts target different pathways simultaneously therefore any solution in-cluding these products may be a smart thought for better results. Manygroups are working in this direction, and the outcomes are promisingtowards the improvement of new helpful cure. In this review, we willexamine the biology of the pancreatic tumor, diagnosis, treatment tech-niques and clinical trials. We will likewise concentrate on the plausiblerole of herbal products, alone or in combination with systemic chemo-preventive medications, in the treatment of the pancreatic tumor.

2. Biology of pancreatic cancer

The biology of pancreatic cancer is perplexing and inadequatelycaught on. Pancreas has both exocrine and endocrine cells that canstructure tumors; however, the likelihood is more for exocrine cells.The vast majority of the exocrine tumors are adenocarcinomas thatbegin in organ cells in the ductal epithelium and advances from prema-lignant injuries to the entirely invasive tumor. Tumors of the endocrinepancreas, commonly termed as islet cell tumors or neuroendocrinetumors, are less common and can be characterized into gastrinomas,insulinomas, glucagonomas, somatostatinomas, VIPomas, PPomas andso forth.

Themicroenvironment of the pancreatic tumor is made out of a fewcomponents, for example, pancreatic cancer cells, pancreatic cancerstem cells (pancreatic CSCs), and the thick, ineffectively vascularizedstroma. The studies suggest that the stroma likewise regulate the

Page 3: BBA Rev on Cancer

Fig. 1.Generation of cancer cells by various cell types. Somatic cells can be converted to progenitor cells which give rise to various types of differentiated cells. Cancer stem cells, progenitorcells, and cells with oncogenic mutations may lead to formation of tumor mass.

15D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

pancreatic tumor growth and development, intrusion and metastasisseparated from its movement as the mechanical boundary. These stro-mal cells consistently interface with cancer cells by different autocrineand paracrine secretion of the pancreas, for example, platelet-derivedgrowth factor (PDGF), transforming growth factor β (TGF-β) andcytokines [19]. These development variables fortify a critical segmentof stroma called pancreatic stellate cells, which in turn express α–smooth-muscle actin and produce rich collagen fibers. These fibersadd to tumor hypoxia (a primary stimulator of tumor movement andmetastasis by influencing angiogenesis), cell survival, and apoptoticpathways [19]. Hypoxia is likewise proposed to be a major cause fordrug/therapy resistance in different tumors [20,21].

Pancreatic tumor cells apparently grow around a population of can-cer stem cells (CSCs) that have the capability of self-renewal andmulti-lineage differentiation [22] (Fig. 1). Pancreatic CSCs can be isolated byflow cytometry utilizing CD44, CD24, and ESA as surface markers [23].Although CD44+/CD24+/ESA+ cells constitute only 0.2-0.8% of thetotal cell population, they are capable of forming tumor spheres [24].Other markers for pancreatic CSCs are CD133, CXCR4, c-Met andALDH1 [25–28]. CXCR4 assumes an essential part in the tumor invasionand metastasis and the cells positive for both CD133 and CXCR4, showhigher metastatic potential than other populations [26,29]. In a recentstudy, it has been demonstrated that human PDACs contain CSCs withhigh levels of CXCR4 and ABCB1, and such patients had reduced survivalrate [30]. Recently Shankar et al. effectively demonstrated the tumori-genic potential of pancreatic CSCs isolated from human pancreatictumors in NOD/SCID mice utilizing surface markers CD44, ESA, CD133,and CD24 [10]. These cells were highly tumorigenic and were likewiseexpressing ALDH and pluripotency maintaining factor, Oct-4 [10].They further indicated high expression of CD133, CD24, CD44, ESA,Nanog, Notch1, MDR1 and ABCG2 in these CSCs (CD133+CD44+CD24+

ESA+ cell population) contrasted with CD133−CD44−CD24−ESA− cellpopulation [10].

Various elements regulate the properties and conduct of pancreaticCSCs, for example, nestin can balance attack or metastasis of pancreatic

CSCs, Oct-4 andNanog can direct pancreatic CSC's conduct andmetastasisto different organs, DCLK1 can segregate between normal and tumoralstem cells and Sox2 controls cell proliferation and differentiation [31].Furthermore, c-Kit and Kras likewise tweak the movement of pancreaticadenocarcinoma [32]. Epithelial tomesenchymal transition (EMT), a pro-cess of change of epithelial attributes into mesenchymal properties, is anurgent process for tumor progression and is proposed to be in charge forthe appearance of cells with stem cell-like properties [33].

CSCs are thought to be themajor contributory element to the absenceof effective treatment for pancreatic malignancy and are in charge of tu-morigenesis, metastasis, and development of chemo and radioresistance[24]. A recent study exhibited that PANC-1 cancer cell line, steadily over-expressing Oct4 and Nanog, show chemoresistance, multiplication, relo-cation, intrusion, and tumorigenesis in vitro and in vivo [34]. Moreover,the ALDH+CD44+CD24+cell population iswell reported to be impervi-ous to treatment with gemcitabine [35]. Removal of CSCs is essentialfor robust tumor treatment, as CSCs stay untouched. Hence, drugs thatcan specifically target CSCs could be a superior alternative for pancreaticcancer prevention.

Pancreatic cancer is hereditarily complex and heterogeneous innature. Different malady conditions, for example, pancreatitis, cysticfibrosis, and inflammation have their effect on the initiation of pancre-atic cancer and its malignant progression [36,37]. Mutations in fourcritical genes namely Kras2, Cdkn2a, TP53 and Dpc4 (or Smad4) are fre-quently seen in pancreatic malignancy patients. The vastmajority of thecancer patients carry one ormore of thesemutations [38]. The observedfrequency of Kras mutation is more than 90%, however, the rate of inac-tivation mutation of Cdkn2a, TP53, and Dpc4 are 95%, 75% and 50%respectively [39,40]. The mutation in Kras2 brings about the consistentexpression of irregular Ras protein that causes aberrant activation of cellproliferation and survival pathways [19]. With increasing age, the like-lihood of acquiring activating mutations in Kras2 gene increases in themajor organs such as lung, pancreas, colon, and other tissues. On theother hand Cdkn2a, TP53 and Dpc4 are the tumor suppressors and mu-tations in these genes result in their inactivation, which facilitate the

Page 4: BBA Rev on Cancer

16 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

proliferation and survival signaling [19,41]. Moreover, a recent studycalled attention to the loss of functionmutation in SWI/SNF nucleosomeremodeling complex in 23% pancreatic adenocarcinomas [42].

3. Signaling pathways in pancreatic cancer

Genetic mutations serve as the basis for abbrent signaling pathways.A comprehensive study with more than 24 pancreatic cancer cases, anaverage of 63 relevent genetic abnormalities (mainly point mutations)per tumor were classified as likely to be relevent in its pathogenesis[43] (reviewed in [19]). Thesemutations can be clubbed in 12 notewor-thy signaling pathways including STAT3, Smad/TGF-β, Wnt, Notch,PI3K/Akt, sonic hedgehog and so forth [43] (reviewed in [19]). Aberrantsignaling in these cellular events has been implicated in the develop-ment and progression of pancreatic tumors by permitting increasedproliferation, angiogenesis, survival, and metastasis (Fig. 2).

4. STAT3 pathway

Signal transducer and activator of transcription 3 (STAT3), encodedby STAT3 gene, is activated in a wide variety of signaling pathways.It mediates diverse responses, including transmission of the signals ofcytokines and growth factors from the cell membrane to the nucleusto regulate gene expression for cell development, differentiation, prolif-eration, survival, and angiogenesis [44–46] (Fig. 3). A range of cytokinesincluding interleukin (IL)-6, IL-9, IL-10, IL-27, tumor necrosis factor α(TNF-α), and monocyte chemotactic protein-1 (MCP-1) activate theSTAT3 pathway [47–49]. Various growth factors, for example, epidermalgrowth factor (EGF), platelet-derived growth factor (PDGF), granulocytecolony-stimulating factor (G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF) also activate this pathway [44–49]. Primaryfunctions of activated STAT3pathway incorporate cell proliferationbyup-regulating cyclin D1 and cyclin B1 and inhibition of apoptosis by up-regulating Bcl-2, Bcl-xL, Mcl-1. Initiated STAT3 assumes a discriminatingpart in tumorigenesis by controlling angiogenesis (VEGF, FGF, andHIF1α), invasion and metastasis (MMP2, MMP9, TWIST, and ICAM1)[44–49].

STAT3 activation has been detected in diverse type of malignancies,and its inhibition by use of inhibitors or short interfering RNA has

Fig. 2.Molecular targets of cancer preventive agents. Cancer preventive agents regulates varioumodulating the expression of various genes.

prompted to reverse the malignant phenotype. STAT3 activation hasbeen portrayed in almost 70% of solid and hematological malignancies[50]. Studies in conditional knockout mice have demonstrated thatSTAT3 pathway is latent in typical pancreas and is not needed for anyvital process related to pancreatic development and homeostasis [51].Nevertheless, it is constitutively activated in PDAC by phosphorylationof Tyr705 in human tumor specimens and also in various PDAC celllines [52–55]. Further, STAT3 is necessary for the development of ADMprocess (acinar-to-ductal metaplasia), which is an early event in PDACpathogenesis mediated by ectopic expression of the Pdx1 [54]. It is re-markable that Pdx1 is a transcription factior and a key regulator ofearly pancreatic development [54]. Another study showed that withmalignant transformation, activated STAT3 promotes proliferation ofcells by regulating G1/S-phase progression and supports the malignantphenotype of human pancreatic cancer [52]. IL-6 signaling dependentactivation of STAT3 plays an important role in promoting PanIN pro-gression and thePDACdevelopment, in addition to oncogenic KrasG12Dtransformation [56]. Themyeloid cells in the pancreas induce STAT3 ac-tivation by releasing IL-6, which promote PanIN progression and thePDAC development.

5. Smad/TGF-β pathway

The transforming growth factor beta (TGF-β) signalingpathway reg-ulates various cellular processes like cell growth, cell differentiation, ap-optosis and cellular homeostasis in both the adult organism and thedeveloping embryo [57]. TGF-β signaling occurs from the membraneto the nucleus via Smad proteins [58]. Smads can be classified into 3major groups; receptor-regulated Smads (R-Smad; Smad1, Smad2,Smad3, Smad5 and Smad8/9), common-mediator Smad (co-Smad;Smad4) and agonistic or inhibitory Smads (I-Smad; Smad6 andSmad7) [58]. Cascade is triggered by the binding of TGF-β superfamilyligand to a type II receptor which catalyzes the recruitment and phos-phorylation of a type I receptor. Subsequently R-Smads are phosphory-lated, linkedwith the coSmad andother factors, andfinally accumulatedin the nucleus to regulate the target gene expression. TGF-β receptor ac-tivation results in Smad2 and Smad3 phosphorylation, which then formheteromeric complexes with Smad4. Furthermore, Smad6 and Smad7,

s cell signaling pathways, cell cycle, cell viability, apoptosis, angiogenesis andmetastasis by

Page 5: BBA Rev on Cancer

Fig. 3. STAT3 signaling pathway. Activation of STAT3 signaling pathway regulates invasion and metastasis, angiogenesis, cell metabolism, inflammation, cell survival and proliferationthrough modulation of several genes.

17D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

can prevent TGF-β signaling by interacting either with the receptor orwith Smad2 and Smad3 [58].

TGF-β signaling pathway impairment because of inactivated Smad4(DPC4) is often recognized in pancreatic carcinomas [59,60]. Jonsonet al., investigated a series of pancreatic carcinoma cell lines with re-spect to alterations of five Smad genes involved in TGF-β signaling,and demonstrated the structural rearrangement of Smad4 in 42% ofthese tumor cells [59]. Since, this pathway could likewise be influencedby other factors that regulate the activation of TGF-β and its receptorgenes, they further assessed expression of uPA, uPAR, IGF2R, TGF-βR1-3,ENG, ALK1, TGF-β1-3, mutations of TGF-βR1-2, cell surface localizationof TGF-βR2 and ENG, and TGF-β1 response in 14 pancreatic carcinomacell lines [59]. The study suggested ALK5- Smad4 as a major target for in-activation in pancreatic carcinomas and that the expression of TGF-βR2,TGF-βR3, and receptors involved in TGF-β activation aremaintained [59].

6. Wnt pathway

Wnt signaling pathway is a complex process and plays an importantrole in tumor development apart from its involvement in other physio-logical and pathological processes [61]. In canonical pathway, the ligandbinding to its receptor (Frizzled/LRP receptor complexes) triggers a cas-cade of events that prevents β-catenin degradation inside the cytoplasmand permits its stabilization and translocation in nucleus where it bindsto transcriptional factors of the Tcf/Lef family forming an activator com-plex. AbnormalWnt/β-catenin signaling is reported in pancreatic cancer[62]. Activated Wnt signal leads to the accumulation of β-catenin in thenucleus where it activates specific target genes [63]. The accumulationof β-catenin is observed both in nucleus and cytoplasm in pancreaticcancer [61,64–66]. The functional evidences are also accumulating thatimplicate a supporting role for β-catenin in PDACmaintenance and pro-gression [61]. It has been also found that β-catenin accumulation and

signaling could be increased through paracrine signaling taking placein the PDAC micro-environment [61]. In a recent investigation, it hasbeen found that Wnt/β-catenin signaling inhibition by wnt-c59 re-sults in reversal of TSA sensitivity, migration ability, and the EMTphenotype in trichostatin A-resistant Panc-1 cells (Panc-1/TSA) [67].

In spite of the fact that our understanding of the role of this pathwayhas increased during the last decade, the general mechanism by whichβ-catenin accumulation occurs in PDAC is poorly understood and needsfurther elucidation.

7. Notch pathway

Notch signaling is shown to regulate proliferation and apoptosisevents in various cell types. The alterations in Notch signaling have vari-ous consequences including tumorigenesis [68,69]. In mammals, fourNotch receptors (Notch1-4) and five ligands (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 sig-naling prompting the cleavage of Notch through a cascade of proteolyticcleavages by themetalloprotease, tumor necrosis factor-α-converting en-zyme (TACE) and γ-secretase complexes [68,71]. The cleavage by TACEgenerates Notch extracellular truncation (NEXT) which is subsequentlycleaved by the γ-secretase complex releasing the active fragment Notchintracellular domain (NICD) from the plasma membrane. NICD translo-cates into the nucleus where it binds tomembers of the CSL transcriptionfactor 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 signalingpathway by Notch1 siRNA or gamma-secretase inhibitors, such as,MRK-003, MK-0752 etc., enhances chemosensitivity to gemcitabinein pancreatic cancer cells through activating apoptosis [73]. Abelet al. reported the significance of Notch pathway in maintaining

Page 6: BBA Rev on Cancer

18 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

cancer stem cell population in pancreatic cancer and investigated theconnection of Notch pathway and percentage of the CSCs population.The inhibition of this pathway resulted in a reduced percentage ofCSCs and tumor sphere formation; notwithstanding, activation dem-onstrated the inverse impact [74]. Further, Lee et al. suggested thatthe activation of the Notch pathway and the increase in CSCs mightcontribute to the failure of treatment in pancreatic cancer [75].Notch has also been shown to be associated with the EMT in pancre-atic cancer [72,76]. It is remarkable that during the EMT process,epithelial cells gain the mesenchymal over endothelial characteristicthereby increasing in migratory and invasive capacity, leading to in-vasion and metastasis [77,78].

8. PI3K/Akt pathway

PI3K pathway acts via phosphorylation of FoxO proteins via Akt,which in turn impairs the DNA-binding ability and increases its affinityfor 14-3-3 proteins [79]. These complexes are exported from the nucle-us to cytoplasm leading to the inhibition of FoxO-mediated survivalpathways. Some other downstream effectors that regulate cell cyclearrest and apoptosis, such as, active FKHRL1, FKHR, and AFX are alsotranslocated to the cytoplasm by similar mechanism [79]. Downstreameffectors of the PI3K-Akt pathway are actively involved in a variety ofvital and specialized functions such as differentiation and proliferationin diversified cells including adipocytes, hepatocytes, myoblasts, thy-mocytes and cancer cells [79].

PI3K/Akt pathway is activated in a variety of cells including fibro-blastic, epithelial, and neuronal cells as survival signal. An elevatedlevel of Akt has been reported in many types of tumors. Studies haveshown the requirement of activated PI3K-Akt/FoxO signaling for thegrowth and survival of the pancreatic tumor. It has been found thatthe 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 reversetranscriptase subunit, FoxOs and IkappaB kinases and so forth [79].

9. Sonic Hedgehog pathway

Sonic hedgehog (Shh) is a member of the Hedgehog (Hh) family ofsecreted signaling proteins. Shh signaling is triggered by binding of thesecreted Shh peptide to Patched (Ptch), which leads to inhibition ofPtch activity. Consequently, Smoothened (Smo) gets phosphorylatedresulting in the activation of the Gli family of zinc-finger transcription fac-tors and therefore target gene expression [80]. Shh signaling has diversefunctions during vertebrate development and post-embryonically in tis-sue homeostasis [81,82]. Alteration in this pathway have been linked tovarious tumor types including pancreatic cancer [81,82]. Activation ofShh signaling pathway has been reported to be involved in the regulationof the pancreatic CSC's expansion,whereas its inhibition (by impairing Glibinding to its promoters) has been demonstrated to upregulate DRs andFas expression, curb Bcl-2 and PDGFRα expressions, and encourage apo-ptotic cell death in pancreatic CSCs [24]. Further, its inhibition has beenshown to reduce tumor-associated stromal tissues, enhance gemcitabineuptake in tumor cells and prolong the average survival rate in pancreaticcancer mouse model [83]. Recently, Rodova et al. showed that inhibitionof Shh pathway components, Gli transcriptional activity and its down-stream targets by sulforaphane inhibited human pancreatic CSCs derivedspheres and induced apoptosis by inhibition of Bcl-2 and activation ofcaspases in vitro [84]. Further, Li et al. showed that inhibition of Shh path-way 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 improvethe outcomes of patients with this devastating disease [85].

10. Treatment of pancreatic cancer: chemotherapy and radiotherapy

The essential choice for pancreatic cancer treatment is its surgicalremoval. However, in advance metastatic stages, the treatment mainlyaims to increase the survival by optimal control of metastases. Systemicchemotherapy may be utilized at any phase of pancreatic malignancywith the objective to minimize the patient's disease-related symptomsand to prolong survival. Presently, a limited number of drugs are acces-sible for the treatment of the pancreatic tumor. Gemcitabine has beenthe reference regimen since 1997 when it was indicated better than 5-Fluorouracil (5-FU) in a phase III clinical trial [86]. A significant concernwith 5-FU was the associated toxicity with its treatment, in particular,gastrointestinal toxicity. A combination of 5-FU and Fluoropyrimidine(S-1) could likewise be a decent choice as S-1 potentiates the antitumoractivity of 5-FU and decreases gastrointestinal toxicity in pancreatictumor mouse models [87]. In recent years different combinations, forexample, the systemic treatment with Abraxane and Gemcitabineand the multidrug combination, FOLFIRINOX, have been attempted[88–109]. Despite the fact that FOLFIRINOX has been promising, it canhave a remarkable reaction profile, constraining its utility in patientswith poor baseline performance status [110].

Radiation therapy, combined with chemotherapy, may be used inpatients whose cancers have grown beyond the pancreas and cannotbe removed by surgery. Uses of radiation treatment alongside high-energy x-beams to kill cancer cells is extremely regular for the treat-ment of advanced stages of tumors. Pancreatic neuroendocrine tumors(NETs) usually do not respond to radiation, and therefore it is rarelyused to treat these tumors. However, it can be used in case of pancreaticNETs that have spread to the bone and other tissues. Furthermore, thetypical radiation treatment utilized for the treatment of the exocrinepancreatic malignancies is External Shaft Radiation Treatment that fo-cuses the radiation on the cancer from a machine outside the body. Ra-diation treatment is typically associated with different side effects, likeskin changes in areas getting radiation, nausea and vomiting, diarrhea,fatigue, poor appetite, weight loss, lower blood counts and last but notthe least increased risk of severe infection.

11. Clinical trials in pancreatic cancer

The chemotherapy drug, Gemcitabine, has been a standard initialtreatment for patients with metastatic pancreatic cancer for over 15years [86]. Various clinical trials have tried new medications, eitheralone or in combination with Gemcitabine (Table 1); however, the ad-vancement is moderate amid the most recent decade [88–109,111].Gemcitabine alone or in combination with Capecitabine or Erlotinibremained the favored systemic treatment alternatives until 2010[88–109]. Since 2010, use of FOLFIRINOX has increased both inmetasta-tic and locally advanced cancer [110,111]. Various drugs and combina-tions have been evaluated in the last couple of years, and some ofthem have shown promising results. Sunitinib and Everolimus haveshown significant improvement in survival. Sunitinib treatmentshowed median progression-free survival of 11.4 months as comparedwith 5.5 months for patients who received the placebo. On the otherhand, the patients receiving Everolimus showed median progression-free survival of 11 months as compared with 4.6 months for patientswho received the placebo. Although these drugs have shown prom-ising results, severe side effects as anemia and neutropenia werealso observed.

Combination of Nab-Paclitaxel (Abraxane®), a form of the che-motherapy drug Paclitaxel bound to the human protein albuminand contained in nanoparticles, and Gemcitabine (Gemzar®) wasalso investigated in an international randomized phase III trial show-ing improved survival [112]. Patients who received the drug combi-nation had a median overall survival of 8.5 months, compared with6.7 months for patients treated with gemcitabine alone [112]. FDAapproved therapy to treat patients with metastatic pancreatic cancer

Page 7: BBA Rev on Cancer

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

Rank Interventions NCT Number Enrollment Conditions

1 Drug: RAD001 NCT00409292 33 Pancreatic Cancer2 Drug: Gemcitabine|Drug: Albumin-bound paclitaxel NCT00398086 67 Metastatic Pancreatic Cancer3 Drug: gemcitabine hydrochloride|Drug: imatinib mesylate NCT00161213 44 Pancreatic Cancer4 Dietary Supplement: genistein|Drug: erlotinib hydrochloride|Drug:

gemcitabine hydrochlorideNCT00376948 20 Pancreatic Cancer

5 Drug: Ixabepilone|Drug: Cetuximab NCT00383149 58 Metastatic Pancreatic Cancer6 Biological: cetuximab|Drug: docetaxel|Drug: irinotecan hydrochloride NCT00042939 94 Pancreatic Cancer7 Drug: alvocidib|Drug: docetaxel NCT00331682 10 Adenocarcinoma of the Pancreas|Recurrent Pancreatic

Cancer|Stage IV Pancreatic Cancer8 Drug: bortezomib|Drug: carboplatin|Other: laboratory biomarker

analysisNCT00416793 9 Acinar Cell Adenocarcinoma of the Pancreas|Duct Cell

Adenocarcinoma of the Pancreas|Stage IV Pancreatic Cancer9 Drug: bevacizumab [Avastin] NCT01214720 607 Pancreatic Cancer10 Drug: Erlotinib, escalating dose|Drug: Erlotinib, standard

dose|Drug: GemcitabineNCT00652366 467 Pancreatic Cancer

12 Drug: Ipilimumab|Biological: Pancreatic Cancer Vaccine NCT00836407 30 Pancreatic Cancer13 Biological: cixutumumab|Drug: erlotinib hydrochloride|Drug:

gemcitabine hydrochlorideNCT00617708 134 Stage IV Pancreatic Cancer

14 Drug: Gemcitabine|Drug: Erlotinib|Drug: Sorafenib NCT00696696 45 Pancreatic Cancer15 Biological: bevacizumab|Drug: erlotinib hydrochloride|Other:

laboratory biomarker analysisNCT00365144 36 Pancreatic Cancer

16 Drug: Capecitabine|Drug: Docetaxel NCT00290693 45 Pancreatic Cancer17 Drug: Abraxane NCT00691054 20 Pancreatic Cancer18 Drug: gamma-secretase/Notch signaling pathway inhibitor RO4929097 NCT01232829 18 Adenocarcinoma of the Pancreas|Recurrent Pancreatic

Cancer|Stage IV Pancreatic Cancer19 Drug: Lenalidomide|Drug: Gemcitabine NCT00837031 72 Metastatic Pancreatic Cancer20 Drug: sorafenib tosylate|Drug: gemcitabine hydrochloride|Other:

laboratory biomarker analysisNCT00114244 52 Stage IV Pancreatic Cancer

21 Drug: GSK1120212|Drug: Gemcitabine|Drug: Placebo NCT01231581 160 Cancer22 Drug: Gemcitabine|Drug: AG-013736|Drug: Gemcitabine NCT00219557 111 Pancreatic Neoplasms23 Drug: dasatinib|Procedure: laboratory biomarker analysis|Procedure:

physiologic testingNCT00474812 49 Adenocarcinoma of the Pancreas|Recurrent Pancreatic

Cancer|Stage IV Pancreatic Cancer24 Drug: gemcitabine|Drug: placebo|Drug: Erlotinib|Drug: apricoxib NCT00709826 109 Pancreatic Cancer|Metastatic Pancreatic Cancer25 Drug: AG-013736|Drug: Gemcitabine|Drug: Gemcitabine|Drug: placebo NCT00471146 630 Carcinoma, Pancreatic Ductal26 Drug: Albumin-bound paclitaxel|Drug: Gemcitabine NCT00844649 861 Metastatic Pancreatic Cancer27 Drug: cohort 1|Drug: cohort 2|Drug: cohort 3|Drug: cohort 4 NCT00439179 27 Metastatic Pancreatic Cancer28 Drug: gemcitabine hydrochloride|Drug: tanespimycin NCT00577889 21 Adenocarcinoma of the Pancreas|Recurrent Pancreatic

Cancer|Stage IV Pancreatic Cancer29 Drug: PCI-27483|Drug: Gemcitabine NCT01020006 42 Pancreatic Cancer|Ductal Adrenocarcinoma|Exocrine

Pancreatic Cancer30 Drug: imexon in combination with gemcitabine|Drug: imexon

placebo + gemcitabineNCT00637247 142 Pancreatic Neoplasms

31 Drug: CO-1.01|Drug: Gemcitabine NCT01124786 367 Metastatic Pancreatic Adenocarcinoma32 Drug: Cetuximab|Drug: Gemcitabine|Drug: Oxaliplatin|Drug:

Capecitabine|Radiation: RadiotherapyNCT00338039 69 Pancreatic Cancer

33 Drug: Gemcitabine NCT00390182 38 Gastrointestinal Neoplasms|Ovarian Neoplasms34 Biological: DTA-H19 NCT00711997 9 Pancreatic Neoplasms35 Drug: Fentanyl sublingual spray|Drug: Placebo NCT00538850 130 Cancer36 Drug: Avastin|Drug: Tarceva|Radiation: Radiation Therapy NCT00735306 12 Pancreatic Cancer37 Drug: Gemcitabine|Drug: Sunitinib NCT00556049 72 Renal Cell Carcinoma|Neoplasm Metastases38 Drug: Gemcitabine|Drug: Placebo|Drug: Sorafenib NCT00661830 103 Adenocarcinoma39 Drug: bevacizumab [Avastin]|Drug: 5 FU|Drug:

Streptozotocin|Drug: XelodaNCT00448136 83 Neoplasms

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

NCT00113230 25 Rectal Cancer

42 Drug: Sunitinib NCT01121562 12 Pancreatic Neuroendocrine Tumors43 Drug: RAD001|Drug: Octreotide Depot NCT00113360 67 Neuroendocrine Carcinoma|Islet Cell Carcinoma44 Drug: Sunitinib malate|Procedure: Hepatic Artery Embolizations NCT00434109 39 Neuroendocrine Tumor|Islet Cell Tumor45 Other: Cocoa Polyphenols NCT01617603 62 Diabetes Type 2

19D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

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 monoclonal antibody that suppresses connec-tive tissue growth factor), have also shown promising results [113].

12. Herbal products, cancer prevention, and pancreatic cancer

The relation between the consumption of certain dietary herbals anda reduced risk of cancer is becoming evident as many epidemiologicaland 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 anda wide range of dietary constituents show potential biological activities[114]. Studies in this line on cell/animal models and human epidemio-logical trials have shown the potential of dietary polyphenols as anti-carcinogenic agents. The reports have shown that phenolic compoundshave the capability to inhibit the molecular events in the cancer initia-tion, promotion, and progression stages. They may increase the expres-sion of pro-apoptotic components in initiated proliferating cells andthereby prevent or delay tumor development. Although it seems thatphenolic compounds induce apoptosis in a precise manner in cancercells but in some human studies no promising results were obtained.

Page 8: BBA Rev on Cancer

20 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

A Cohort Study of Diet and Cancer in Netherlands suggested no effect ofthe consumption of black tea on the risk for colorectal, stomach, lungand breast cancers [115]. A similar studywasperformed in Japan involv-ing more than 25,000 stomach cancer patients with a similarobservation i.e. no association of consumption of green tea with gastriccancer risk [116]. Someother studies ondifferent types of cancer also in-dicated the same conclusion [117–119]. On the contrary, a decreasedrisk for the different types of cancer has been reported after the con-sumption of flavonoids or certain foods or drinks rich in these phenoliccompounds [118–122].

The use of herbals in the treatment of pancreatic cancer is a novelapproach and is continuously gaining the attention of investigators.Previous research in this area was focused on inducing apoptosis butrecently these herbals have beenused in the targeting of other key path-ways of cell survival, angiogenesis, metastasis, and differentiation. Easyavailability and less or no toxicity even at higher doses have made themthe preferable choice over other cancer chemopreventive options (Fig. 2).

Table 3Summary of recent findings showing chemopreventive potential of curcumin againstpancreatic cancer.

Cells/animals Effect Reference

MIA PaCa-2 and mouse (tumorxenograft model)

Inhibits the proliferation andenhances apoptosis in MIA PaCa-2cells and inhibits tumor growthand the expression of thetranscription nuclear factor NF-κBand NF-κB-regulated geneproducts in xenograft mousemodel

[220]

MiaPaCa-2 and Panc-1 cells Down-regulates the expression ofmiR-221 resulting in upregulationof PTEN, p27(kip1), p57(kip2),and PUMA leading to the

[221]

13. Resveratrol

Resveratrol, a phytoalexin, is commonly found as an ingredient in redwine, skins of grapes, peanuts and so forth. It possess anti-tumorigenic,anti-inflammatory, and anti-oxidant properties [123]. Its preventive roleagainst cancers, cardiovascular diseases, and various neurological disor-ders has been widely reported [124–126]. Hydroxylation of resveratrolby CYP1B1 generates two major metabolites namely piceatannol and3,4,5,4′-tetrahydroxystilbene that substantially contribute to its chemo-preventive activities by inhibiting tyrosine kinase and inducing apoptosis[127–129].

Chemopreventive effects of resveratrol against various cancers havebeen extensively investigated in both in vitro and in vivo. It has beenshown to have anti-tumor activity by inhibiting angiogenesis, endothe-lial cell migration, tumor formation and by blockage of oxygen free rad-ical formation [130–132] (Table 2). Due to its lipophilic nature, it readilycrosses the plasmamembrane and establishes dynamic homeostasis byinhibiting the phase I (mainly CYP450s) and inducing phase II enzymes(UDP-glucuronosyl transferase, NAD(P)H quinone oxidoreductase,and glutathione-s-transferases) during stress conditions [133–138]. Incancerous cells, it inhibits the expression of inducible nitric oxide (NO)synthase and NO production [139]. It also inhibits the formation of apreneoplastic lesion inmousemammary glands and proliferation of a va-riety of cancer cells in culture including, human colon, breast, andprostatecancer cells [140–143].

Resveratrol sensitizes a broad spectrum of tumors including lungcarcinoma, acute myeloid leukemia, promyelocytic leukemia, multiple

Table 2Summary of recent findings showing chemopreventive potential of resveratrol againstpancreatic cancer.

Cells Effect Reference

MIA PaCa-2 cells Inhibits proliferation and induces apoptosis [213]Panc-28 and Hs766Tcells

Increases calcium levels and preventsmigration of TG2-expressing cells

[214]

Capan-2 cells Inhibits tumor growth, induced apoptosis, andup-regulated Bax and VEGF-B expression

[215]

BxPC-3 and Panc-1 Inhibits cell proliferation, migration, andinduces expression of EMT-related genes(E-cadherin, N-cadherin, vimentin, MMP-2,and MMP-9)

[216]

PANC-1, CFPAC-1, andMIA Paca-2 cells

Inhibits viability and miR-21 expression andincreases Bcl2 expression

[217]

BxPC-3 and Panc-1 Inhibits the growth Gli1, Ptc1, CCND1, andBCL-2

[218]

PANC-1, MIA PaCa-2,Hs766T, and AsPC-1

Up-regulates p21/CIP1, p27/KIP1, Bim,activates csapase-3, reduces phosphorylationof ERK, PI3K, Akt, FoxO1, and FoxO3a

[219]

myeloma, prostate cancer, oral epidermoid carcinoma, and pancreaticcancer. Zhou et al. showed that it enhances caspase-3 activationand p53 and p21 expression in capan-2 and colo357 pancreatic cancercell lines [144]. A recent approach, using human pancreatic CSCs(CD133+CD44+CD24+ESA+), showed that resveratrol sensitizes andinhibits the growth and development of pancreatic cancer lesion inKrasG12D mice [10]. This study further showed that the resveratrolinhibits pluripotency maintaining factors (Nanog, Sox-2, c-Myc and Oct-4), drug resistance gene ABCG2, CSC's migration, invasion, self-renewal,and components of EMT (Zeb-1, Slug, and Snail) [10]. Resveratrol inhibitscell growth, proliferation and expression of the anti-apoptotic proteinsBcl-2, Bcl-xL, and XIAP and induces apoptosis, cell cycle arrests, caspasesand pro-apoptotic gene Bax in pancreatic cancer cell lines [145]. Addition-ally, resveratrol has been found to suppress proliferation and anchorage-independent growth of pancreatic cancer by inhibiting leukotriene B4(LTB4) production and expression of the LTB4 receptor 1 (LTB4R1)[146]. It is remarkable that LTB4 is a hydrolysis product of the leukotrieneA4 (LTA4), and the process is catalyzed by LTA4hydrolase, a known targetfor prevention and therapy of cancers including PC [147]. Resveratrol candirectly bind to leukotriene A4 hydrolase (LTA-4H) and inhibit its activityand, therefore, LTB4 production [146].

14. Curcumin

Curcumin is one of themost commonly used and highly investigatedphytochemical. During the last decade, our understanding of its thera-peutic potential and themultiplemechanismsbywhich it offers chemo-prevention against various cancers has been increased [148] (Table 3).The various pharmacological effects of curcumin include apoptotic,anti-proliferative, anti-oxidant, and anti-angiogenic properties. The

inhibition of cell proliferation andmigration

MIA PaCa-2 and mouse (tumorxenograft model)

Inhibits cell proliferation, reducestumor growth and angiogenesis asdetermined by a reduced numberof blood vessels and decreasedexpression of vascular endothelialgrowth factor and annexin A2proteins

[222]

BxPC-3 and MiaPaCa-2 Activates TNFR, CASP 8, CASP3,BID, BAX, and down-regulatesNFκB, NDRG 1, and BCL2L10 gene

[223]

TGF-β1-stimulated PANC-1 cells Inhibits proliferation, inducesapoptosis and reverses the EMT

[224]

AsPC-1 and MiaPaCa-2 Decreases pancreatic cancer cellsurvival, clonogenicity, formationof pancreatospheres, invasive cellmigration, and CSC function

[225]

AsPC-1, MiaPaCa-2, Panc-1,BxPC-3 human and Pan02mouse pancreatic cancer cells

Inhibits tumor growth throughmitotic catastrophe by increasingthe expression of RNA bindingprotein CUGBP2, therebyinhibiting the translation of COX-2and VEGF expression

[151]

Page 9: BBA Rev on Cancer

21D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

previous studies have highlighted that curcumin targets multiple signaltransduction pathways and that it suppresses a number of essentialelements in cellular signaling pathways, for example, phosphorylationcatalyzed by protein kinases, c-Jun-activated protein 1 (AP-1) activationand prostaglandin biosynthesis.

It has also been found that curcumin potentiates radiotherapy in PCcure probably by involving selective regulation of radiotherapy-inducedNF-κB [149]. Studies have shown that curcumin inhibits cell prolifera-tion and induces apoptotic cell death mediated by PARP cleavage andCaspase-3 in MIAPaCa-2, Panc-1 and BxPC-3 pancreatic cancer cells[150]. Subramaniam et al. showed a significant reduction in tumor vol-ume and angiogenesis in curcumin treated tumor xenografts [151].They further showed that curcumin inhibits cell proliferation, inducesof G2-M arrest and apoptosis, enhances phosphorylation of checkpointkinase 2 (Chk2) coupled with higher levels of nuclear cyclin B1 andCdc-2, and increases expression of cyclooxygenase-2 (COX-2) [151].Curcumin also inhibits ERK activity and suppresses EGFR andNotch-1 sig-naling leading to increased apoptosis in pancreatic cancer. Glienke et al.showed that incubation with curcumin results in down-regulation ofWilms' tumor gene 1 (WT1; a gene frequently expressed in pancreaticcancer) in a dose-dependent manner [152]. Additionally, curcumin hasbeen shown to restrain STAT3 and induce apoptosis by inhibiting theexpression of the anti-apoptotic gene Survivin/BIRC4 in pancreatic cancercells [153].

Recently Bar-Sela et al. reviewed the accomplished and continuingclinical trials with curcumin as an anticancer agent [154]. In one trial,17 patients were treated with the oral dose of 8 gm/day of curcuminin combination with Gemcitabine. Although the results showed thatthis combined treatment is tolerable in patients; nevertheless, it hasbeen suggested to reduce the dose of curcumin [155]. Dhillon et al.used only curcumin as the 1st line treatment for the 25 patients. Theyfound that curcumin down regulates the expression of NFκB, COX-2,and the phosphorylation of STAT3 in peripheral blood [156]. In spite ofthese encouraging results, extensive clinical trials are needed beforedrawing any conclusion.

15. Epigallocatechin gallate (EGCG)

Epigallocatechin gallate (EGCG) is a most extensively studied cate-chin and the major polyphenol present in green tea. Various studieshave shown that EGCG offers protection against pancreatic canceramong the other tumors (Table 4); however, the exactmolecularmech-anismbywhich EGCG suppresses humanpancreatic cancer cell prolifer-ation is unclear. Kürbitz et al. showed anticancer properties of EGCG onhuman pancreatic ductal adenocarcinoma (PDAC) cells PancTu-I, Panc1,Panc89 and BxPC3 in vitro [157]. They found that EGCG inhibits prolifer-ation of PDAC cells in a dose- and time-dependent manner. The proteinexpression analysis performed with PancTu-I cells evidently showedEGCG-mediated modulation of cell cycle regulatory proteins (cyclins,

Table 4Summary of recent findings showing chemopreventive potential of EGCG against pancreatic ca

Cells/animals Effect

PANC-1 Suppresses proliferation and induces apopAsPC-1 cells Regulates RKIP/ERK/NF-κB and/or RKIP/NBalb c nude mice (tumor xenograft model) Inhibits pancreatic cancer orthotopic tumo

pathways and activation of FKHRL1/FoxO3Colo357 human pancreatic adenocarcinomacells

With PGHS-2-specific inhibitor celecoxib,down-regulates release of pro-angiogenicmatrix metalloproteinase (MMP)-2

PANC-1 Inhibits HIF-1α protein expression, P-gp mAsPC-1 and PANC-1 Suppresses the growth, invasion, and migr

and enhances the therapeutic potential ofHuman pancreatic cancer stem cells(CD133+/CD44+/CD24+/ESA+)

Inhibits Nanog, c-Myc and Oct-4 expressio(smoothened, patched, Gli1 and Gli2) andXIAP and activating caspase-3

cyclin-dependent kinases, and inhibitors). The study further statedthat EGCG inhibits TNFα-induced activation of NF-κB and consequentlysecretion of pro-inflammatory and invasion-promoting proteins likeIL-8 and uPA [157]. Moreover, previous studies have demonstratedthat EGCG decreases cell adhesion ability on micro-pattern dots, ac-companied by dephosphorylation of both focal adhesion kinase andinsulin-like growth factor-1 receptor (IGF-1R) in AsPC-1 and BxPC-3 cells [158,159,64]. EGCG has been also found to aid retained activa-tion 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. Astudy by Li Y et al. showed that the binding of EGCG to Hsp90 impairsthe association of Hsp90 with its co-chaperones, thereby inducingdegradation of Hsp90 client proteins (Akt, Cdk4, Raf-1, Her-2, andpERK) consequently anti-proliferating effects in pancreatic cancer cells[160]. Basu and Haldar showed that EGCG causes the disappearance ofintact 21 kDa Bid protein and induces activation of caspase-8 leadingto cell death in MIA PaCa-2 cells [161]. Further, involvement of trans-membrane extrinsic signaling in this polyphenol triggered pancreaticcarcinoma cell death was confirmed by RNase protection assay thatclearly showed up-regulation of the members of death receptor family[161,160].

Shankar et al. examined the role of EGCG in inhibiting growth, inva-sion, metastasis and angiogenesis of human pancreatic cancer cells in axenograft model system [162]. They found that EGCG inhibits viability,capillary tube formation, and migration of HUVEC. Additionally, theyobserved EGCG-mediated inhibition of proliferation (Ki-67 and PCNAstaining), angiogenesis (vWF, VEGF and CD31) and metastasis (MMP-2, MMP-7, MMP-9 and MMP-12), and induction of apoptosis (TUNEL),caspase-3 activity and growth arrest (p21/WAF1) in vivo [162]. Theyalso found a significant reduction in the circulating vascular endothelialgrowth factor receptor 2 (VEGF-R2) positive endothelial cells, ERK ac-tivity, and induction of p38 and JNK activities in vivo following EGCGtreatment [162].

16. Genistein

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

ncer.

Reference

tosis, modulates the PI3K/Akt/mTOR signaling pathway [226]F-κB/Snail and inhibits invasive metastasis [227]r growth, angiogenesis, and metastasis, inhibits PI3K/Akt and ERKa

[162]

synergistically diminishes metabolic activity via apoptosis induction andvascular endothelial growth factor (VEGF) and invasiveness-promoting

[228]

RNA and protein levels and cell proliferation [229]ation, induces apoptosis by interfering with the STAT3 signaling pathwaygemcitabine and CP690550

[230]

n, self-renewal, proliferation, EMT, components of Shh pathwayGli transcriptional activity, and induces apoptosis by inhibiting Bcl-2 and

[231]

Page 10: BBA Rev on Cancer

Table 5Summary of recent findings showing chemopreventive potential of sulforaphane againstpancreatic cancer.

Cells/animals Effect Reference

Established BxPc-3 and AsPC-1 PDAcell lines and immortalizedCRL-4023 hTERT-HPNE humanPDA cells

Increases Cx43 and E-cadherinlevels, inhibits c-Met andCD133, improved thefunctional morphology andcommunication of gapjunctions.

[232]

MIA PaCa-2 and Panc-1 Inhibits cell viability and NF-κBDNA binding activity, inducescell apoptosis by activation ofcaspase-3 and PARP cleavage,increases pERK1/2, c-Jun, p38MAPK, p53 protein expressionwhen used in combinationwith aspirin and curcumin

[233]

NOD/SCID/IL2Rgamma mice (tumorxenograft model)

Inhibits growth of tumors,expression of Shh pathwaycomponents, EMT,pluripotency maintainingtranscription factors,angiogenic markers andinduces apoptosis

[85]

Human normal pancreatic stem cells(HPSC) and human pancreaticcancer stem cells(CD133+/CD44+/CD24+/ESA+)

Inhibits CSC's derived spheres,components of Shh pathwayand Gli transcriptional activity,expression of pluripotencymaintaining factors (Nanogand Oct-4) as well as PDGFRαand Cyclin D1

[84]

Pancreatic cancer xenograft mousemodel

Disrupts protein-proteininteraction in Hsp90 complexfor its chemopreventiveactivity

[234]

MIA-PaCa2 Potentiates the inhibitoryeffects of gemcitabine and5-flurouracil on clonogenicity,spheroid formation, ALDH1activity, Notch-1 and c-Relexpression

[235]

PANC-1, MIA PaCa-2 and AsPC-1 Inhibits cell proliferation,colony formation,phosphorylation of Akt andERK, activates FoxOtranscription factors andinduces apoptosis

[79]

22 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

effect of genistein onDNA topoisomerase II is also amajor contributor toits cytotoxic activity [170,172].

Genistein inhibits cell growth, clonogenicity, cell migration and in-vasion, EMT phenotype, and formation of pancreatospheres consistentwith reduced expression of CD44 and EpCAM [185].Wang et al., showedthat genistein restricts pancreatic cancer cell invasion by inhibiting cellgrowth and inducing apoptosis alongwith attenuation of FoxM1 and itsdownstream genes (survivin, Cdc25a, MMP-9, and VEGF) [186].

17. Sulforaphane

Sulforaphane has been reported to inhibit the growth of establishedtumors and prevent chemically induced cancers in animal models[187–189] (Table 5). It has been shown to inhibit Akt pathway in ovar-ian, prostate and colorectal cancers [189–191] and down-regulateβ-catenin in HeLa and HepG2 cells [192]. Additionally, it also targetsbreast cancer stem/progenitor cells effectively in both in vitro andin 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, whereascontrol tumor cells quickly generate large tumors [194].

Furthermore, sulforaphane has also been shown to inhibit theself-renewal capacity of pancreatic CSCs. Srivastava et al., showed thatinhibition of Nanog enhances the inhibitory effects of sulforaphane on

the self-renewal capacity of CSCs [195]. Sulforaphane induces apoptosisby activating caspase-3 and inhibiting the expression of Bcl-2 and XIAP,as well as phosphorylation of FKHR. Additionally, sulforaphane issuggested to block signaling involved in early metastasis by inhibitingthe expression of proteins involved in the epithelial-mesenchymal tran-sition (beta-catenin, vimentin, TWIST1, and ZEB1) [195]. Additionally,sulforaphane, in combination of with TRAIL, has been suggested to bea promising strategy for targeting pancreatic tumor initiating cells(TICs). It has been found that it could abrogate the resistance of pancre-atic TICs to TRAIL (tumor necrosis factor-related apoptosis-inducing li-gand) by interfering with TRAIL-activated NF-κB signaling [196].

18. Garlic

Garlic has been traditionally used for varied human ailments aroundthe world. Epidemiological observations and preclinical studies, both incell and animal models, suggest the anti-carcinogenic potential of garlicand its constituents [197]. Chemical analysis revealed that the protec-tive effects of garlic are due to the presence of organosulfur compoundsmainly allyl derivatives [197]. Additionally, it modulates the activity ofseveralmetabolizing enzymes involved in the activation and detoxifica-tion of carcinogens and inhibits DNA adduct formation. It possessanti-oxidative and free radicals scavenging properties and regulatescell proliferation, apoptosis, and immune responses. Recent data sug-gest that garlic also modulates cell-signaling pathways to avoid prolifera-tion of unwanted cells thereby imparting strong cancer chemopreventive,as well as cancer therapeutic effects [197].

19. Benzyl isothiocyanate (BITC)

Due to their capability to induce apoptosis, modulate signalingpathways and inhibit angiogenesis, isothiocyanates (ITCs) have showna great promise as chemopreventive agents against various tumors inrecent years [198]. Benzyl isothiocyanate (BITC) is a major ITC com-pound present in cruciferous vegetables. BITC suppresses the initiationand progression of a variety of cancers including lung, esophageal,forestomach, urinary bladder, mammary, liver, colon, and pancreatictumors [198–203]. Various preclinical and mechanistic studies havesupported the anticancer efficacy of BITC, and it has been found to sup-press the growth of human pancreatic cancer cells both in vitro andin vivo. BITC is reported to induceG(2)/Mphase cell cycle arrest, and ap-optotic cell death in pancreatic cancer cell/animal models [204–207].The apoptotic potential of BITC is attributed to its capability to activateMAPK family members i.e. ERK, JNK and P38 by catalyzing their phos-phorylation at Thr202/Tyr204, Thr183/Tyr185, and Thr180/Tyr182respectively in a dose-dependent manner [206]. Additionally, the po-tential to inhibit the phosphorylation and expression of NF-kB mostlikely via inhibition of HDAC1/HDAC3, is suggested to be another con-tributory factor to the apoptotic potential of BITC [204].

Furthermore, it has been found to inhibit angiogenesis andmetasta-sis by suppressing VEGF and MMP-2 expression in pancreatic cancercells [208]. Recently Boreddy et al., showed that BITC offers protectionagainst pancreatic tumor growth by effectively containing STAT-3 andHIF-1α and VEGF expression in BxPC-3 and PanC-1 pancreatic cancercells [208]. It reduces the phosphorylation of PI3K, Akt, Pdk1, mTOR,FoxO1, and FoxO3a and increases apoptosis in tumor xenograft mousemodel. BITC treatment also decreases the binding of FoxO1 with 14-3-3protein suggesting it's nuclear retention and subsequent elevationof FoxO-responsive proteins involved in apoptosis (Bim) and cell cyclearrest (p27 and p21) [209].

BITC has also been shown to sensitize pancreatic tumors forradiotherapy. BITC treatment in a combination of X-rays or gamma-irradiation reduces cell survival as compared with individual (X-raysor gamma-irradiation) exposure in pancreatic cancer cells. This effectis suggested to be due to the inhibition of cell proliferation and anti-apoptotic genes like XIAP/IAP, and augmentation of apoptosis protease

Page 11: BBA Rev on Cancer

23D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

activating factor-1 (Apaf-1) triggered by BITC [208]. It is remarkablethat Apaf-1 is essential for activation of caspase-9 in stress-inducedapoptosis [208].

20. Piperlongumine

Piperlongumine (PL) is an alkaloid found in the fruits of long pepperplants that displays potent growth-inhibitory properties in a varietyof cancer cell lines and various animal models. It has been identifiedto target cancer cells selectively over normal cells through an ROS-dependent mechanism in a cell-based small-molecule screening andquantitative proteomics approach [210]. It increases ROS levels andcancer-selective cell death by directly binding and inhibiting the antiox-idant enzyme glutathione S-transferase pi 1 (GSTP1) [210,211]. Rajet 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 sug-gested that PL also targets BxPC-3 pancreatic cancer cells that containwild-type K-ras [212]. They further showed the anti-cancer effects ofPL in vivo. PL reduces tumor volume, increases oxidative DNA damage(8-OHdGhigh), and reduces proliferation (Ki-67low) in nude mice xeno-grafts for PANC-1 [212].

21. Conclusion and future perspectives

Pancreatic cancer is continuously posing a challenge to the cliniciansand researchers. We are still relying on the old traditional therapies.The major drawback of current therapy is their unilateral actions onone or two pathways whereas the approach should be to target severalevents simultaneously. The combination therapy is a right approach inthis direction, but associated side effects are a major concern. Sincethe small population of pancreatic CSCs is mostly responsible for thepathogenesis of pancreatic cancer, an efficient therapy targeted to pan-creatic CSCs is also an excellent approach. However, the problem is theresistance of pancreatic CSCs against conventional treatment but stilldevelopmental pathways such as the hedgehog, Wnt, Notch, etc. canbe targeted.

Flavonoids have emerged as potential chemopreventive candidatesfor cancer treatment, especially by their ability to induce apoptosis.These can interfere with the initiation, development and progressionof cancer by the modulation of cellular proliferation, differentiation,apoptosis, angiogenesis, and metastasis. Flavonoids have been shownto target cancer cells specifically with no or insignificant effects onhealthy cells in vitro. Nevertheless, some studies suggest to includeexperimental conditions (dose, cell type, culture conditions and treat-ment length) while interpreting the results of in vitro studies becausethe biological outcome can be affected. Since the apparent phenomenonis a result of complex interaction of different cellular events, the mech-anisms for inducing the apoptosis of these polyphenols may overlapwith other signaling cascades. Therefore, using polyphenols to promoteprogrammed cell death through the modulation of different proteins inother pathways that can contribute to apoptosis could be an alternativeapproach.

In summary, the characteristic effects of flavonoids, such as, inductionof apoptosis, activation of caspases, down-regulation or up-regulation ofBcl-2 family members, induction of cell cycle arrest and inhibition of sur-vival/proliferation make them ideal therapeutic candidate for pancreaticcancer prevention. Nevertheless, although the results from in vitroexperiments constitute a valuable tool for elucidating the pathwaysinvolved in the overall carcinogenesis process, these cannot be directlyextrapolated to clinical effects. Despite the fact that the herbal productstarget multiple signaling events simultaneously, more studies will beneeded to understand clearly the mechanisms of their action and thera-peutic potential.

Disclosure of potential conflicts of interest

The authors have declared that no Conflicts of Interest exist.

Acknowledgments

We acknowledge our lab members for critical reading of the manu-script, insightful discussions, and valuable advice. The project was fundedby the National Institutes of Health (RKS) and The VA Merit Award (SS).

References

[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 adenocarcinomaand pancreatic intraepithelial neoplasia: an immunohistochemical analysis withautomated 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 dysfunctionof 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 carci-noma PANC-1 cells Can Act as cancer vaccine to enhance antitumor immuneresponse in mice, Oncol. Res. 21 (2013) 307–316.

[4] S.M. Conrath, The use of epidemiology, scientific data, and regulatory authority todetermine risk factors in cancers of some organs of the digestive system. 6. Pancre-atic 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, Pancre-atic cancer: progress in cancer therapy, Crit. Rev. Oncol. Hematol. 67 (2008) 27–38.

[7] A. Neesse, S. Krug, T.M. Gress, D.A. Tuveson, P. Michl, Emerging concepts in pancre-atic cancer medicine: targeting the tumor stroma, Onco Targets Ther. 7 (2013)33–43.

[8] M. Bardou, I. Le Ray, Treatment of pancreatic cancer: A narrative review of cost-effectiveness studies. Best practice & research, Clin. Gastroenterol. 27 (2013)881–892.

[9] T. Kolodecik, C. Shugrue, M. Ashat, E.C. Thrower, Risk factors for pancreatic cancer:underlying mechanisms and potential targets, Front. Physiol. 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 andKrasG12D transgenic mice by inhibiting pluripotency maintaining factorsand epithelial-mesenchymal 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 futureprospects, 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.

[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 stemcells, 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 Res. 67(2007) 1030–1037.

[24] C.J. Lee, J. Dosch, D.M. Simeone, Pancreatic cancer stem cells, J. Clin. Oncol. Off. J.Am. Soc. Clin. Oncol. 26 (2008) 2806–2812.

[25] M. Herreros-Villanueva, A. Zubia-Olascoaga, L. Bujanda, c-Met in pancreatic cancerstem cells: therapeutic implications, World J. Gastroenterol. 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 andmetastatic 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, Establish-ment and characterization of a highly tumourigenic and cancer stem cell enrichedpancreatic cancer cell line as a well defined model system, PLoS One 7 (2012)e48503.

Page 12: BBA Rev on Cancer

24 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

[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 activ-ity of gemcitabine by modulation of c-Met pathway in pancreatic cancer, Curr.Pharm. Des. 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, Highlylymphatic metastatic pancreatic cancer cells possess stem cell-like properties, Int.J. Oncol. 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 containsa 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 per-spectives on cancer therapeutics, World J. Gastroenterol. 20 (2014) 10790–10801.

[32] A. Amsterdam, C. Raanan, N. Polin, E. Melzer, D. Givol, L. Schreiber, Modulation ofc-kit expression in pancreatic adenocarcinoma: a novel stem cell marker responsi-ble for the progression of the disease, Acta Histochem. 116 (2014) 197–203.

[33] J.A. Castellanos, N.B. Merchant, N.S. Nagathihalli, Emerging targets in pancreaticcancer: epithelial-mesenchymal transition and cancer stem cells, Onco TargetsTher. 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 expres-sion inhibits the stemness of pancreatic cancer cells, Cancer Lett. 340 (2013)113–123.

[35] S. Yabuuchi, S.G. Pai, N.R. Campbell, R.F. deWilde, E. De Oliveira, P. Korangath,M.M.Streppel, Z.A. Rasheed, M. Hidalgo, A. Maitra, N.V. Rajeshkumar, Notch signalingpathway targeted therapy suppresses tumor progression and metastatic spreadin pancreatic cancer, Cancer Lett. 335 (2013) 41–51.

[36] P. Maisonneuve, B.C. Marshall, A.B. Lowenfels, Risk of pancreatic cancer in patientswith cystic fibrosis, Gut 56 (2007) 1327–1328.

[37] H. Yu, D. Pardoll, R. Jove, STATs in cancer inflammation and immunity: a leadingrole for STAT3, Nat. Rev. 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 pathwaysin pancreatic carcinoma, Cancer Res. 57 (1997) 1731–1734.

[41] G. Feldmann, R. Beaty, R.H. Hruban, A. Maitra, Molecular genetics of pancreaticintraepithelial neoplasia, J. Hepatobiliary Pancreat. Surg. 14 (2007) 224–232.

[42] A.H. Shain, J.R. Pollack, The spectrum of SWI/SNF mutations, ubiquitous in humancancers, 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 pathwaysin human pancreatic cancers revealed by global genomic analyses, Science 321(2008) 1801–1806.

[44] N.K. Sharma, S. Shankar, R.K. Srivastava, STAT3 as an emerging molecular target inpancreatic cancer, Gastrointestinal Cancer: Targets and Therapy 2014 (4) (2014)115–122.

[45] M. Bar-Natan, E.A. Nelson, M. Xiang, D.A. Frank, STAT signaling in the pathogenesisand treatment of myeloid malignancies, JAKSTAT 1 (2012) 55–64.

[46] P. Yue, J. Turkson, Targeting STAT3 in cancer: how successful are we? Expert Opin.Investig. Drugs 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 pathogenesisand treatment of myeloid malignancies, JAKSTAT 1 (2012) 55–64.

[49] P. Yue, J. Turkson, Targeting STAT3 in cancer: how successful are we? Expert Opin.Investig. Drugs 18 (2009) 45–56.

[50] A.K. Mankan, F.R. Greten, Inhibiting signal transducer and activator of transcription3: rationality and rationale design of inhibitors, Expert Opin. Investig. Drugs 20(2011) 1263–1275.

[51] M.S. Chang, H.S. Lee, B.L. Lee, Y.T. Kim, J.S. Lee, W.H. Kim, Differential proteinexpression between esophageal squamous cell carcinoma and dysplasia, and prog-nostic 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 transcrip-tion 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 Januskinase/signal transducer and activator of transcription-3 pathway inhibits growthof human pancreatic cancer, Cancer Lett. 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 pancreascauses acinar-to-ductal metaplasia through Stat3 activation, Genes Dev. 20 (2006)1435–1440.

[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 endothelialgrowth factor and human pancreatic cancer angiogenesis and metastasis, Onco-gene 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/Socs3activation by IL-6 transsignaling promotes progression of pancreaticintraepithelial neoplasia and development of pancreatic cancer, Cancer Cell19 (2011) 456–469.

[57] Y.G. Chen, A.M. Meng, Negative regulation of TGF-beta signaling in development,Cell Res. 14 (2004) 441–449.

[58] Y. Shi, J. Massague, Mechanisms of TGF-beta signaling from cell membrane to thenucleus, 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 inpancreatic carcinomas, Int. J. Oncol. 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 distinctexpression responses to TGFB1, Genes Chromosom. Cancer 36 (2003) 340–352.

[61] J.P.t. Morris, S.C. Wang, M. Hebrok, KRAS, Hedgehog,Wnt and the twisted develop-mental 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 andbeta-catenin expression changes in pancreatic intraepithelial neoplasia, Clin.Cancer Res. 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/beta-catenin 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 signalingin 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, Rever-sion of trichostatin A resistance via inhibition of the Wnt signaling pathway inhuman pancreatic cancer cells, Oncol. Rep. 32 (5) (2014) 2015–2022.

[68] Z. Wang, Y. Li, F.H. Sarkar, Notch signaling proteins: legitimate targets for cancertherapy, Curr. Protein Pept. Sci. 11 (2010) 398–408.

[69] Z. Wang, Y. Li, S. Banerjee, F.H. Sarkar, Exploitation of the Notch signaling pathwayas a novel target for cancer therapy, Anticancer Res. 28 (2008) 3621–3630.

[70] L. Miele, H. Miao, B.J. Nickoloff, NOTCH signaling as a novel cancer therapeutictarget, Curr. 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 pancreaticcancer progression, Pancreat. Disord. Ther. 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 togemcitabine 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 inmaintaining the cancer stem cellpopulation in pancreatic cancer, PLoS One 9 (2014) e91983.

[75] J.Y. Lee, S.Y. Song, J.Y. Park, Notch pathway activation is associated with pancreaticcancer treatment failure, Pancreatology 14 (2014) 48–53.

[76] Z. Wang, Y. Li, D. Kong, F.H. Sarkar, The role of Notch signaling pathway inepithelial-mesenchymal transition (EMT) during development and tumor aggres-siveness, Curr. Drug Targets 11 (2010) 745–751.

[77] B. De Craene, G. Berx, Regulatory networks defining EMT during cancer initiationand progression, Nat. Rev. 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 dissem-ination 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 path-ways causes activation of FOXO transcription factor, leading to cell cycle arrestand apoptosis in pancreatic cancer, J. Mol. Signal. 5 (2010) 10.

[80] T. Osterlund, P. Kogerman, Hedgehog signalling: how to get from Smo to Ci and Gli,Trends Cell Biol. 16 (2006) 176–180.

[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 incarcinogenesis, 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, Hedgehoginhibition prolongs survival in a genetically engineeredmousemodel of pancreaticcancer, Gut 57 (2008) 1420–1430.

[84] M. Rodova, J. Fu, D.N. Watkins, R.K. Srivastava, S. Shankar, Sonic hedgehogsignaling inhibition provides opportunities for targeted therapy by sulforaph-ane 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 Sonichedgehog-GLI pathway, Mol. Cell. Biochem. 373 (2013) 217–227.

[86] H.A. Burris III, 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 clinicalbenefit with gemcitabine as first-line therapy for patients with advanced

Page 13: BBA Rev on Cancer

25D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

pancreas cancer: a randomized trial, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 15(1997) 2403–2413.

[87] M. Fukushima, [Antitumor activity and function of S-1, a new oral tegafur-based for-mulation], Gan to kagaku ryoho, Cancer Chemother. 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 ofexatecan and gemcitabine compared with gemcitabine alone in untreated advancedpancreatic cancer, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 24 (2006) 4441–4447.

[89] J. Berlin, P. Catalano, J. Thomas, J. Kugler, D. Haller, A. Benson, E.C.O.G.T. E, Phase IIIstudy of gemcitabine in combination with fluorouracil versus gemcitabine alone inpatients with advanced pancreatic carcinoma, J. Clin. Oncol. Off. J. Am. Soc. Clin.Oncol. 20 (2002) 3270–3275.

[90] S. Bramhall, J. Schulz, J. Nemunaitis, P. Brown, M. Baillet, J. Buckels, A double-blindplacebo-controlled, randomised study comparing gemcitabine and Marimastatwith gemcitabine and placebo as first line therapy in patients with advancedpancreatic 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 pluscisplatin compared with single-agent gemcitabine as first-line treatment ofpatients with advanced pancreatic cancer: the GIP-1 study, J. Clin. Oncol. Off. J.Am. Soc. Clin. Oncol. 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 capecit-abine in patients with advanced pancreatic cancer, J. Clin. Oncol. Off. J. Am. Soc.Clin. Oncol. 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 cisplatincompared with gemcitabine alone in advanced pancreatic cancer, J. Clin. Oncol.Off. J. Am. Soc. Clin. Oncol. 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 capecitabineplus erlotinib followed by gemcitabine in advanced pancreatic cancer: final resultsof 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, Gemcitabineplus capecitabine compared with gemcitabine alone in advanced pancreaticcancer: a randomized, multicenter, phase III trial of the swiss group for clinicalcancer Res and the central european cooperative oncology group, J. Clin. Oncol.Off. J. Am. Soc. Clin. Oncol. 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 gemcitabinein patients with advanced pancreatic adenocarcinoma: a double-blind randomisedphase 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 inlocally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCADphase III trial, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 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. Voskoglou-Nomikos, M.Ptasynski, W. Parulekar, Erlotinib plus gemcitabine compared with gemcitabinealone in patients with advanced pancreatic cancer: a phase III trial of the NationalCancer Institute of Canada Clinical Trials Group, J. Clin. Oncol. Off. J. Am. Soc. Clin.Oncol. 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 ofpemetrexed plus gemcitabine versus gemcitabine in patients with unresectableor 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. Abbruzzese,C. Blanke, Phase III study comparing gemcitabine plus cetuximab versusgemcitabine in patients with advanced pancreatic adenocarcinoma: SouthwestOncology Group- directed intergroup trial S0205, J. Clin. Oncol. Off. J. Am. Soc.Clin. Oncol. 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 gemcitabineoxaliplatin (GEMOX) in patients with advanced pancreatic cancer (E6201), J.Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 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 pancreaticadenocarcinoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 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, fluoroura-cil, and gemcitabine in advanced pancreatic cancer: a randomised controlledmulticentre 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, multi-center, phase III trial of gemcitabine, 5-fluorouracil (5- FU), folinic acid vs.gemcitabine alone in patients with advanced pancreatic cancer, J. Clin. Oncol. Off.J. Am. Soc. Clin. Oncol. 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 survivaladvantage compared with gemcitabine monotherapy in patients with locallyadvanced or metastatic pancreatic cancer despite increased tumor responserate, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 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- dosegemcitabine alone or in combination with capecitabine in patients with metastaticpancreatic 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 comparingirinotecan-gemcitabine (IG) with gemcitabine (G)monotherapy as first-line treat-ment in patients with locally advanced or metastatic pancreatic cancer, Br. J. Can-cer 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 randomisedphase III trial comparing gemcitabine with surgery-only in patients with resectedpancreatic cancer: Japanese Study Group of Adjuvant Therapy for PancreaticCancer, 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 withgemcitabine plus placebo in advanced pancreatic cancer, J. Clin. Oncol. Off. J. Am.Soc. Clin. Oncol. 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, N. Engl. J. Med.364 (2011) 1817–1825.

[111] D. Papadatos-Pastos, K. Thillai, R. Rabbie, P. Ross, D. Sarker, FOLFIRINOX – a newparadigm in the treatment of pancreatic cancer, Expert. Rev. Anticancer. Ther. 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 withnab-paclitaxel plus gemcitabine, N. Engl. J. Med. 369 (2013) 1691–1703.

[113] E. Yu, M.W. Saif, K. Huber, Phase 1 trials in pancreatic cancer, J. Pancreas 15 (2014)326–328.

[114] S. Ramos, Effects of dietary flavonoids on apoptotic pathways related to cancerchemoprevention, 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, N.Engl. J. Med. 344 (2001) 632–636.

[117] R. Garcia, C.A. Gonzalez, A. Agudo, E. Riboli, High intake of specific carotenoids andflavonoids does not reduce the risk of bladder cancer, Nutr. Cancer 35 (1999)212–214.

[118] I.C. Arts, P.C. Hollman, H.B. Bueno De Mesquita, E.J. Feskens, D. Kromhout, Dietarycatechins and epithelial cancer incidence: the Zutphen elderly study, Int. J. Cancer92 (2001) 298–302.

[119] R. Garcia-Closas, A. Agudo, C.A. Gonzalez, E. Riboli, Intake of specific carotenoidsand 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 ofcancer 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 amonga Japanese population, Prev. Med. 26 (1997) 769–775.

[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 epithelialcells: molecular mechanisms, Am. J. Physiol. Lung Cell. Mol. Physiol. 287(2004) L774–L783.

[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 clinicalstudies, Anticancer Res. 24 (2004) 2783–2840.

[125] L.M. Hung, J.K. Chen, S.S. Huang, R.S. Lee, M.J. Su, Cardioprotective effect of resver-atrol, a natural antioxidant derived from grapes, Cardiovasc. Res. 47 (2000)549–555.

[126] Z.J. Sun, C.E. Pan, H.S. Liu, G.J. Wang, Anti-hepatoma activity of resveratrol in vitro,World J. Gastroenterol. 8 (2002) 79–81.

Page 14: BBA Rev on Cancer

26 D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

[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 agentresveratrol is converted to the anticancer agent piceatannol by the cytochromeP450 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, Involvementof cytochrome P450 1A2 in the biotransformation of trans-resveratrol in humanliver 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 thegrowth 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 cyto-chrome P450 enzymes by epsilon-viniferin, the dimer of resveratrol: comparisonwith resveratrol and polyphenols from alcoholized beverages, Life Sci. 73 (2003)1199–1213.

[131] B. Piver, F. Berthou, Y. Dreano, D. Lucas, Inhibition of CYP3A, CYP1A and CYP2E1activities 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 vascularendothelial cadherin tyrosine phosphorylation, Mol. Pharmacol. 64 (2003)1029–1036.

[133] G. Upadhyay, A.K. Singh, A. Kumar, O. Prakash, M.P. Singh, Resveratrol modulatespyrogallol-induced changes in hepatic toxicity markers, xenobiotic metabolizingenzymes 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, Liquidchromatography/tandem mass spectrometric determination of inhibition ofhuman 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 CYP1A1in vitro by preventing activation of the aryl hydrocarbon receptor, Cancer Res. 58(1998) 5707–5712.

[136] T.K. Chang, J. Chen, W.B. Lee, Differential inhibition and inactivation of humanCYP1 enzymes by trans-resveratrol: evidence for mechanism-based inactivationof CYP1A2, J. Pharmacol. Exp. Ther. 299 (2001) 874–882.

[137] H. Szaefer, M. Cichocki, D. Brauze, W. Baer-Dubowska, Alteration in phase I and IIenzyme activities and polycyclic aromatic hydrocarbons-DNA adduct formationby 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-cellchronic 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 chemopre-ventive 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 resveratrolon human breast epithelial cells, Int. J. Oncol. 12 (1998) 865–869.

[142] S.H. Mitchell, W. Zhu, C.Y. Young, Resveratrol inhibits the expression and functionof the androgen receptor in LNCaP prostate cancer cells, Cancer Res. 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 andwine, on human colonic cancer cells, Cancer Lett. 158 (2000) 85–91.

[144] J.H. Zhou, H.Y. Cheng, Z.Q. Yu, D.W. He, Z. Pan, D.T. Yang, Resveratrol induces apo-ptosis 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 andcolon 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, ared wine polyphenol, suppresses pancreatic cancer by inhibiting leukotrieneAhydrolase, Cancer Res. 70 (2010) 9755–9764.

[147] X. Chen, S. Wang, N. Wu, C.S. Yang, Leukotriene A4 hydrolase as a target for cancerprevention and therapy, Curr. Cancer Drug Targets 4 (2004) 267–283.

[148] A. Shehzad, F. Wahid, Y.S. Lee, Curcumin in cancer chemoprevention: moleculartargets, pharmacokinetics, bioavailability, and clinical trials, Arch. Pharm.(Weinheim, Ger.) 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 onRel protein-regulated cell death/radiosensitization in pancreatic cancer cells,Pancreas 40 (7) (2011) 1107–1119.

[150] S. Aggarwal, M.W. Ndinguri, R. Solipuram, N. Wakamatsu, R.P. Hammer, D. Ingram,W. Hansel, [DLys(6)]-luteinizing hormone releasing hormone-curcumin conjugateinhibits pancreatic cancer cell growth in vitro and in vivo, Int. J. Cancer 129 (2011)1611–1623.

[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 mediatescurcumin-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 atarget 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 STAT3phosphorylation in human pancreatic cancer cell lines and downregulationof survivin/BIRC5 gene expression, Cancer Investig. 28 (2010) 166–171.

[154] G. Bar-Sela, R. Epelbaum, M. Schaffer, Curcumin as an anti-cancer agent: re-view 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 gemcitabinein patients with advanced pancreatic cancer, Nutr. Cancer 62 (2010) 1137–1141.

[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 patientswith advanced pancreatic cancer, Clin. Cancer Res. 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 epigallocate-chin gallate in growth suppression and anti-inflammatory activities in pancreatictumor 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 antican-cer effect in human pancreatic carcinoma cells via the inhibition of both focaladhesion 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-3-gallateinhibits Hsp90 function by impairing Hsp90 associationwith cochaperones in pancre-atic 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 onpancreatic cancer cell death, Int. J. Oncol. 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) 440–452.

[163] P.B. Kaufman, J.A. Duke, H. Brielmann, J. Boik, J.E. Hoyt, A comparative survey ofleguminous plants as sources of the isoflavones, genistein and daidzein: implica-tions 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, Chemopreventionof colon carcinogenesis by dietary administration of piroxicam, alpha-difluoromethylornithine, 16 alpha-fluoro-5-androsten-17-one, and ellagicacid individually and in combination, Cancer Res. 51 (1991) 4528–4534.

[165] R.C. Alves, I.M. Almeida, S. Casal, M.B. Oliveira, Isoflavones in coffee: influence ofspecies, roast degree, and brewing method, J. Agric. Food Chem. 58 (2010)3002–3007.

[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, Peroxisomeproliferator-activated receptor gamma (PPARgamma) as a molecular target forthe 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. vander Burg, J.A. Gustafsson, Interaction of estrogenic chemicals and phytoestrogenswith estrogen receptor beta, Endocrinology 139 (1998) 4252–4263.

[170] M. Lopez-Lazaro, E. Willmore, C.A. Austin, Cells lacking DNA topoisomerase II betaare 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 Nrf2-Keap1defense pathway in cardiovascular disease: protection afforded by dietaryisoflavones, 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 kinaseinhibitor 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 combina-tion of indol-3-carbinol and genistein synergistically induces apoptosis in humancolon cancer HT-29 cells by inhibiting Akt phosphorylation and progression ofautophagy, 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 anddehydroascorbic 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 DNAstrand breakage in human HL-60 and K-562 leukemia cells by genistein, CancerRes. 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 relateddifferences in susceptibility of HL-60 cells to apoptosis induced by various antitu-mor 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 humanmyeloid leukemiacell line associated with the tyrosine-phosphorylation and activation of the proto-oncogene c-kit product, Blood 78 (1991) 2834–2840.

[179] B.Q. Li, M. Subleski, D. Shalloway, H.F. Kung, T. Kamata, Mitogenic activation of theRas guanine nucleotide exchange factor in NIH 3T3 cells involves protein tyrosinephosphorylation, Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 8504–8508.

[180] C. Linassier, M. Pierre, J.B. Le Pecq, J. Pierre, Mechanisms of action in NIH-3T3 cellsof genistein, an inhibitor of EGF receptor tyrosine kinase activity, Biochem.Pharmacol. 39 (1990) 187–193.

Page 15: BBA Rev on Cancer

27D. Singh et al. / Biochimica et Biophysica Acta 1856 (2015) 13–27

[181] D. Linnekin, L.S. Park, W.L. Farrar, Dissociation of human cytokine receptor expres-sion 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 topo-isomerase 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 onthe growth and cell cycle progression of normal human lymphocytes and humanleukemic 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 andcancer stem cell phenotype in pancreatic cancer cells, J. Cell. Biochem. 112 (2011)2296–2306.

[186] Z. Wang, A. Ahmad, S. Banerjee, A. Azmi, D. Kong, Y. Li, F.H. Sarkar, FoxM1 is anovel 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 ofinducers of enzymes that protect against chemical carcinogens, Proc. Natl. Acad.Sci. U. S. A. 94 (1997) 10367–10372.

[188] Y. Zhang, T.W. Kensler, C.G. Cho, G.H. Posner, P. Talalay, Anticarcinogenic activitiesof sulforaphane and structurally related synthetic norbornyl isothiocyanates, Proc.Natl. Acad. Sci. U. S. A. 91 (1994) 3147–3150.

[189] F.L. Chung, C.C. Conaway, C.V. Rao, B.S. Reddy, Chemoprevention of colonicaberrant 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 bynatural dietary compounds sulforaphane and dibenzoylmethane alone and incombination in ApcMin/+ mouse, Cancer Res. 67 (2007) 9937–9944.

[191] D. Chaudhuri, S. Orsulic, B.T. Ashok, Antiproliferative activity of sulforaphane inAkt-overexpressing ovarian cancer cells, Mol. Cancer Ther. 6 (2007) 334–345.

[192] S.Y. Park, G.Y. Kim, S.J. Bae, Y.H. Yoo, Y.H. Choi, Induction of apoptosis by isothiocy-anate sulforaphane in human cervical carcinoma HeLa and hepatocarcinomaHepG2 cells through activation of caspase-3, Oncol. Rep. 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/broccolisprouts, inhibits breast cancer stem cells, Clin. Cancer Res. 16 (2010) 2580–2590.

[194] Y. Li, T. Zhang, S.J. Schwartz, D. Sun, Sulforaphane potentiates the efficacy of17-allylamino 17-demethoxygeldanamycin against pancreatic cancer throughenhanced 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 synergizeswith quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells,Front. Biosci. 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 pancreatictumour-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 Lett. 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 carcinogenmetabolism, 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 ben-zyl 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 tocarcinogen exposure, Cancer Res. 41 (1981) 2991–2994.

[203] Y. Zhang, P. Talalay, C.G. Cho, G.H. Posner, A major inducer of anticarcinogenicprotective 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 isothiocyanate-mediatedinhibition of histone deacetylase leads to NF-kappaB turnoff in human pancreaticcarcinoma 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 pancreaticcancer 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 isothiocyanate-mediatedgeneration of reactive oxygen species causes cell cycle arrest and induces apoptosisvia activation of MAPK in human pancreatic cancer cells, Carcinogenesis 30 (2009)1744–1753.

[207] R. Zhang, S. Loganathan, I. Humphreys, S.K. Srivastava, Benzyl isothiocyanate-induced DNA damage causes G2/M cell cycle arrest and apoptosis in humanpancreatic cancer cells, J. Nutr. 136 (2006) 2728–2734.

[208] S.R. Boreddy, R.P. Sahu, S.K. Srivastava, Benzyl isothiocyanate suppresses pancreat-ic tumor angiogenesis and invasion by inhibiting HIF-alpha/VEGF/Rho-GTPases:pivotal role of STAT-3, PLoS One 6 (2011) e25799.

[209] S.R. Boreddy, K.C. Pramanik, S.K. Srivastava, Pancreatic tumor suppression bybenzyl 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, 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 signal-ing 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 celldeath by enhancing reactive oxygen species and DNA damage, Toxic. Rep. 1(2014) 309–318.

[213] Y. Qin, Z. Ma, X. Dang, W. Li, Q. Ma, Effect of resveratrol on proliferation andapoptosis of human pancreatic cancer MIA PaCa-2 cells may involve inhibition ofthe Hedgehog signaling pathway, Mol. Med. Rep. 10 (2014) 2563–2567.

[214] A. Kumar, J. Hu, H.A. LaVoie, K.B. Walsh, D.J. DiPette, U.S. Singh, Conformationalchanges 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, J. Cancer Res.Clin. Oncol. 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 cancercells via suppression of the PI-3K/Akt/NF-kappaB pathway, Curr. Med. Chem. 20(2013) 4185–4194.

[217] P. Liu, H. Liang, Q. Xia, P. Li, H. Kong, P. Lei, S. Wang, Z. Tu, Resveratrol inducesapoptosis of pancreatic cancers cells by inhibiting miR-21 regulation of BCL-2expression, Clin. Transl. Oncol. 15 (2013) 741–746.

[218] W. Mo, X. Xu, L. Xu, F. Wang, A. Ke, X. Wang, C. Guo, Resveratrol inhibits prolifer-ation and induces apoptosis through the hedgehog signaling pathway in pancreat-ic cancer cell, Pancreatology 11 (2011) 601–609.

[219] S.K. Roy, Q. Chen, J. Fu, S. Shankar, R.K. Srivastava, Resveratrol inhibits growth oforthotopic 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 inhibitstumor growth and angiogenesis in an orthotopic mouse model of human pancre-atic cancer, Biomed Res. Int. 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 cellsthrough up-regulation of PTEN, p27(kip1), p57(kip2), and PUMA, Am. J. CancerRes. 3 (2013) 465–477.

[222] A.P. Ranjan, A.Mukerjee, L. Helson, R. Gupta, J.K. Vishwanatha, Efficacy of liposomalcurcumin in a human pancreatic tumor xenograft model: inhibition of tumorgrowth and angiogenesis, Anticancer Res. 33 (2013) 3603–3609.

[223] M. Youns, G.M. Fathy, Upregulation of extrinsic apoptotic pathway in curcumin-mediated antiproliferative effect on human pancreatic carcinogenesis, J. Cell.Biochem. 114 (2013) 2654–2665.

[224] X.D. Sun, X.E. Liu, D.S. Huang, Curcumin reverses the epithelial-mesenchymaltransition of pancreatic cancer cells by inhibiting the Hedgehog signaling pathway,Oncol. Rep. 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 byswitching on suppressor microRNAs and attenuating EZH2 expression, CancerRes. 72 (2012) 335–345.

[226] S. Liu, X.J. Wang, Y. Liu, Y.F. Cui, PI3K/AKT/mTOR signaling is involved in (−)-epi-gallocatechin-3-gallate-induced apoptosis of human pancreatic carcinoma cells,Am. J. Chin. Med. 41 (2013) 629–642.

[227] S.O. Kim, M.R. Kim, (−)-Epigallocatechin 3-gallate inhibits invasion by inducingthe expression of Raf kinase inhibitor protein in AsPC1 human pancreatic adeno-carcinoma cells through the modulation of histone deacetylase activity, Int. J.Oncol. 42 (2013) 349–358.

[228] C. Hardtner, G. Multhoff, W. Falk, J. Radons, (−)-Epigallocatechin-3-gallate, a greentea-derived catechin, synergizes with celecoxib to inhibit IL-1-induced tumorigen-ic mediators by human pancreatic adenocarcinoma cells Colo357, Eur. J.Pharmacol. 684 (2012) 36–43.

[229] Z. Zhu, Y. Wang, Z. Liu, F. Wang, Q. Zhao, Inhibitory effects of epigallocatechin-3-gallate on cell proliferation and the expression of HIF-1alpha and P-gp in thehuman pancreatic carcinoma cell line PANC-1, Oncol. Rep. 27 (2012) 1567–1572.

[230] S.N. Tang, J. Fu, S. Shankar, R.K. Srivastava, EGCG enhances the therapeutic poten-tial of gemcitabine and CP690550 by inhibiting STAT3 signaling pathway in humanpancreatic cancer, PLoS One 7 (2012) e31067.

[231] S.N. Tang, J. Fu, D. Nall, M. Rodova, S. Shankar, R.K. Srivastava, Inhibition of sonichedgehog pathway and pluripotency maintaining factors regulate human pancre-atic 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 bydysregulated Cx43-mediated gap junctional intercellular communication,Oncotarget 5 (2014) 1621–1634.

[233] A. Thakkar, D. Sutaria, B.K. Grandhi, J. Wang, S. Prabhu, The molecular mechanismof action of aspirin, curcumin and sulforaphane combinations in the chemopreven-tion of pancreatic cancer, Oncol. Rep. 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 cancerthrough disrupting Hsp90-p50(Cdc37) complex and direct interactions withamino 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 to-ward cancer stem-like cells of pancreas and prostate, Mol. Ther. 19 (2011) 188–195.


Recommended