Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2011 Dec 18.
Published in final edited form as: Cancer Prev Res (Phila). 2010 Nov 2;3(11):1382–1387. doi: 10.1158/1940-6207.CAPR-10-0258

Deploying Mouse Models of Pancreatic Cancer for Chemoprevention Studies

Paul J Grippo 1, David A Tuveson 2
PMCID: PMC3242034  EMSID: UKMS32212  PMID: 21045161

Abstract

With the advent of mouse models that recapitulate the cellular and molecular pathology of pancreatic neoplasia and cancer, it is now feasible to recruit and deploy these models for the evaluation of various chemopreventive and/or anticancer regimens. The highly lethal nature of pancreatic ductal adenocarcinoma (PDAC) makes multiple areas of research a priority including assessment of compounds that prevent or suppress the development of early lesions that can transform into PDAC. Currently, there are over a dozen models available, which range from homogeneous preneoplastic lesions with remarkable similarity to human pancreatic intraepithelial neoplasms (PanINs) to models with a more heterogeneous population of lesions including cystic papillary and mucinous lesions. The molecular features of these models may also vary in a manner comparable to the differences observed in lesion morphology, and so navigating the route of model selection is not trivial. Yet, arming the community of cancer investigators with a repertoire of models and the guidance to select relevant models that fit their research themes promises to produce findings that will have clinical relevance.

Introduction

Pancreatic cancer develops insidiously, recurs quickly following surgical resection, and metastasizes widely resulting in nearly uniform lethality. Although grim, these clinical characteristics nonetheless represent several opportunities to interrupt disease progression and improve the outcome for pancreatic cancer patients. Indeed, increased surveillance of individuals with a strong family history of pancreatic cancer has prompted the development of endoscopic, pathological, and radiological methods that allow for highly beneficial prophylactic surgery (1). Furthermore, the limited but measurable benefit of adjuvant chemotherapy supports the premise that systemic treatments can also impact pancreatic cancer progression (2, 3); the challenge, though, is to identify the most effective systemic approaches for different stages of disease. The recent advent of genetically engineered mice that accurately develop early and advanced forms of the most common type of pancreatic cancer, pancreatic ductal adenocarcinoma (PDAC), may provide preclinical model systems to address these issues.

Mouse Models of Pancreatic Cancer for Potential Chemoprevention Studies

Persistent research has culminated in the generation of genetically engineered mouse models that represent different stages of human PDAC. These models are now available to investigate the basic and translational aspects of this malignancy (4, 5). Models of neoplasms such as murine pancreatic intraepithelial neoplasia (mPanIN), intraductal papillary mucinous neoplasia (mIPMN), and mucinous cystic neoplasia (mMCN) have all been described (though mIPMN and mMCN should be further characterized for a more-complete validation). These genetically engineered mouse models may be appropriate for assessing the role of genes, environmental conditions such as tobacco exposure and diet, co-morbidities including pancreatitis, and the influence of immunological and pharmacological interventions on the development of invasive PDAC, as highlighted in Table 1. Models of localized PDAC have also been reported and could be utilized for neoadjuvant, adjuvant, and anti-metastatic approaches to prevent relapse and malignant dissemination. Most of the genetically engineered models include Pdx1-Cre/ Lox-Stop-Lox–responsive (LSL)-Kras or p48Cre/ LSL-Kras mice, which were further modified by conditional deletion or mutation of the p16/p19 (ref. 6; these mice also develop sarcomatoid lesions), p53 (7), smad4 (8, 9), or transforming growth factor β receptor II (TGFβRII) loci (10). The results of these combined genotypes were a multiplicity of preinvasive lesions of all grades, invasive adenocarcinoma, and metastasis to other organs ultimately leading to significantly reduced median survival. The most robust murine models of preneoplasms (earlier-stage lesions, as opposed to later-stage lesions such as PanIN3) display complete penetrance (all mice with a gene mutation have phenotypic manifestation of that disease) and express an endogenous or transgenic oncogenic Kras allele in pancreatic exocrine and/or progenitor cells. When combined with various tumor-suppressor mutations, these models oftentimes yield invasive and metastatic PDAC and related epithelial histologies (see Table 2 in ref. 5). Models using inducible alleles of Cre recombinase, such as estrogen receptor-Cre fusion genes (CreER or CreERT) and tetracycline-responsive Cre expression alleles (TRE-Cre), are capable of being temporally controlled and thus initiated selectively in adult pancreata, better reflecting the somatic acquisition of genetic mutations thought to occur in humans (1115).

Table 1.

Phenotypic comparisons. Similarities and differences among genetically engineered mouse (GEM) models of pancreatic neoplasia for chemoprevention studies

GEM Time of
Express
Preneo/Neoplastic Lesions Mesenchymal Parenchymal Cancer Phenotype Survival
(mos)
Caveats to Consider Ref
mPanIN mIPMN mMCN other AH ADM Fibros Inflam Atrophy Onset Path Inv Met Potential Uses
EL-Kras E.14 +/− + + CPN +++ +++ ++ ++ ++ No
cancer
No No 18+ huKras; driven off EL promoter (69)
Broad screening of mult lesions
Pdx1-Cre
LSL-Kras
E8.5 +++ ++ +++ 6+ PDAC 20% 1
yr
Yes 16 Express in GI tract (22)
Eval effect on PanIN lesions
p48Cre
LSL-Kras
E9.5 +++ ++ +++ 8+ PDAC 20% 1
yr
Yes 16 (22)
Eval effect on PanIN lesions
+Mist1Kras E10.5
+ in
adults
+/− +/− CPN ++ +++ 3 Mixed
histo
Yes Yes 10.8
median
HCC (70)
Broad screening of nonPanIN lesns
Nest-Cre
LSL-Kras
E10.5 ++
1 & 2
(~100%)
+/− ++ +++ No
No No 2 Expression in brain (71)
Eval effect on early PanINs
EL-tTA
TRE-Cre
LSL-Kras
E.16.5
P10
P60
3 mos
5 mos
none
+ ++ +++ +++ ++ 12 PDAC
PDAC
yes
yes
No
No
18+ Kras mt G12V (13)
Chemoprevention prior to express
K5-Cox-2 E13.5 +/− + CN ++ +++ +++ near
PDAC
No No 6-8 10-week=CP;bladder abnorm (24)
Ability to inhibit Cox-2
Pdx1-CreERT
LSL-Kras
E.10.5
P14,21,
24,27,56
2-4 mos
1-2 lower in P56
++ +++ ++ +++ study terminated at 4 mos. addition of R26NIC increase
number & severity of mPanINs
(11)
6 PDAC NR NR term at
6m
Chemoprevention prior to express
EL-CreERT
LSL-Kras
P42 1-30%
2-18%
3-3%
++ ++ ++ No
cancer
No No term at
16 mos.
addition of R26NIC increase
number & severity of mPanINs
(11, 14)
Chemoprevention prior to express
Mist1CreERT
LSL-Kras
P42 2 mos
(like EL
target)
++ No cancer No No term at
12 mos.
Targeting of liver cells (14)
Chemoprevention prior to express
EL-CreERT
cLGL-Kras
E16+ 0-2 = 1,2
2-6 = 1-3
6-9 =inc3
++ +++ ++ ++ 4+ CPC
PDAC
Yes Yes Cre expression without tamoxifen (15)
Chemoprevention prior to express
pCPA1CreERT
LSL-Kras
P14,21,
24,27,56
1 – 10% NR NR NR NR No
cancer
No No term
at 3
mos.
addition of cerulein increase
number & severity of mPanINs
(12)
RipCreERT
LSL-Kras
P14,21,
24,27,56
none No
cancer
No No up to
8 mos
addition of cerulein leads to
development of mPanINs
(12)

Abbreviations: ADM, acinar-ductal metaplasia; AH, acinar hyperplasia; Cox-2, cyclooxygenase-2; CN, cystic neoplasms; CP, chronic pancreatitis; CPC, cystic papillary carcinoma; CPN, cystic papillary neoplasms; EL-Kras, elastase-driven mutant Kras; EL-tTA, EL tetracycline transactivator; Fibros, fibrosis; histo, histology; Inflam, inflammation; Inv, invasive; LSL-Kras, Lox-Stop-Lox-responsive mutant Kras; Met, metastatic; mIPMN, mouse intraductal papillary mucinous neoplasia; Mist1Cre, knock-in of Cre upstream of the Mist1 coding region; Mist1Kras, knock-in of mutant Kras upstream of the Mist 1 coding region; mMCN, mouse mucinous cystic neoplasia; mPanIN, mouse pancreatic intraepithelial neoplasia; NR, not reported; p48Cre, knock-in of Cre upstream of the p48/Ptfa coding region; pCPA1CreERT, procarboxypeptidase A1-responsive CreERT; PDAC, pancreatic ductal adenocarcinoma; RipCreERT, rat insulin promoter-responsive CreERT; TRE-Cre, tetracycline-responsive Cre.

Experimental Design/Strategies and Criteria for Evaluating Interventional Effects

The following pertinent parameters are included among those to consider for pancreatic cancer chemoprevention studies: 1) The optimal in vivo model system for the prevention of invasive cancer or metastasis; 2) criteria for assessing a significant response; and 3) targets of chemoprevention including the relevant molecular pathways, cell types, and environmental conditions promoting the progression of PanIN to PDAC.

Considerations for model selection

Murine models of preneoplasms that progress to invasive PDAC and murine models of focal PDAC that progresses to metastatic disease allow distinct questions to be addressed for cancer prevention and therapy studies (see Table 4.1 in ref. 16). From the perspective of chemoprevention research, perhaps the most critical feature is activation of the target pathway, where the molecular profile of the model is identified prior to the preclinical trial. Timing of delivery may also be a key factor since late administration in models that progress to PDAC may yield only a modest, if any, response. The onset, penetrance, frequency, and latency of progression of these various models should be considered during the design of prevention studies. Some models have a very homogeneous population of neoplastic lesions that may create an ideal environment for studying a single species of lesions. Other models have a variety of lesions that may offer a broader platform for evaluation. Furthermore, the type of preneoplasms (e.g., mPanIN, mIPMN, mMCN) that develop should be considered. These parameters will define the number of mice necessary to achieve adequate statistical power for the analysis, the length of the study, the cellular response, and an indication of the potential mechanisms involved. At times, it may be important to consider the background strain of the mice, the presence of cell-surface antigens, and the source and type of oncogene mutation, particularly as it relates to immunological studies. Other types of lesions and/or abnormalities in the parenchymal and mesenchymal compartment may also play a role, albeit smaller than that of the previously mentioned parameters, in choosing ideal models for preclinical chemoprevention trials and evaluations of the interplay between epithelial and stromal components.

Parameters for assessing response to treatment

A variety of approaches is often used to measure the efficacy of interventions. Direct evaluation of the neoplastic tissue, where onset, incidence, frequency, size, and proliferative/apoptotic indices are assessed, is normally the first point of analysis. This evaluation is often accompanied by molecular evaluations of certain cellular markers and/or factors involved in signaling pathways, especially targets of the chemopreventive agent under assessment. More complex evaluations can include surrogate biomarker investigations in the blood and radiological assessments of cellular/tissue response via small-animal imaging including high-resolution ultrasound and magnetic resonance (17). Such approaches will optimize meaningful analyses of response when performed in tandem.

Targets for chemoprevention

Inflammation

A pivotal question concerning cancer prevention study—for which mouse models are ideally suited—is whether the target is contained in the preneoplastic cells, the microenvironment, or both. This is particularly germane in pancreatic cancer since pancreatitis, which causes both the death of acinar cells and a reactive stromal fibrosis, promotes the development of PDAC in patients carrying the PRSS1 allele (18) and in mice treated with the secretagogue cerulein (12, 13, 1921). Thus far, chemoprevention that suppresses inflammation has been somewhat limited, with a primary focus on elevated levels of cyclooxygenase 2 (COX-2) in mPanIN cells in Pdx1-Cre/LSL-KrasG12D mice (22). Treatment with the nonsteroidal anti-inflammatory drug (NSAID) nimesulide inhibited COX-2 and led to reduced mPanINs, particularly later-stage lesions (mPanIN 3; ref. 23). Similar results were observed in K5-COX-2 transgenic mice (24). Furthermore, a successful preclinical trial of the selective COX-2–inhibiting NSAID celecoxib, a MUC1 peptide, and gemcitabine led to a complete lack of development of invasive disease and significant suppression of mPanINs 2 and 3 in p48Cre/LSL-Kras/MUC1 mice, supporting COX-2 and MUC1 as cancer-chemopreventive targets in the mouse pancreas (25). Complementary genetic approaches to determine whether the EL-PRSS1 allele (19, 21) cooperates with oncogenic Kras in PanIN/PDAC progression, and conversely whether the conditional deletion of COX-2 (26) in pancreatic cells or surrounding stroma inhibits PanIN/PDAC formation, are technically feasible but hitherto unreported.

The nuclear factor kappa B (NF-κB) pathway, a central mediator of inflammatory signaling in neoplastic and microenvironment cells, has been implicated in pancreatic cancer biology (27). Treatment with aspirin as a surrogate pharmacological inhibitor of the NF-κB pathway inhibited orthotopic tumor formation in mice (28, 29). Therefore, more precise inhibition of NF-κB signaling with conditional knock-out alleles or chemical inhibitors of IKB kinases is a logical next step to confirm the relevance of this pathway in PanIN/PDAC. Last, 5-lipoxygenase (5-LOX) is also a reasonable target for many of the same reasons that COX-2 is, namely its presence in early neoplastic lesions (30), its ability to inhibit proliferation and induce apoptosis in culture (31, 32), and its improved in vivo efficacy with gemcitabine (33).

Diet/environment

Obesity, fat and sugar intake, and tobacco exposure have all been implicated in PDAC development and should be evaluated in these neoplastic models. The size and metastatic spread of transplanted Pan02-derived cancers were enhanced in obese Lep(Ob) and Lep(db/db) mice, leading to a significant reduction in survival compared with wild-type mice (34, 35). These findings imply that the obese state may establish an environment conducive to cancer cell proliferation and dissemination, which may also hold true for preneoplastic and neoplastic lesions of the pancreas. Another study maintained p48Cre/LSL-Kras mice on a high-fat diet (HFD) for up to 10 weeks and found greatly increased mPanIN formation that was associated with tumor necrosis factor receptor 1 (TNFR1)-dependent inflammation (36). Elastase-driven (EL)-CreER/LSL-Kras mice had significantly lower fasting blood glucose levels following administration of this HFD (36). A similar approach was employed in the related EL-Kras model, where a diet rich in omega-3 fatty acid significantly reduced the incidence and frequency of pancreatic neoplasms (37). Additional reports propose that high caloric consumption (e.g., high-fructose corn syrup) and/or an increase in glycemic load can have a different means of promoting pancreatic cancer growth (3840), though not without controversy (41). New avenues of research need to determine the molecular and signaling pathways which are influenced by these dietary components (particularly fatty acids and sugars) and may serve as chemoprevention targets. Regarding a metabolic contribution, it appears that diabetes may also serve as a risk factor for PDAC (42).

An environmental factor that markedly increases the incidence of PDAC is cigarette smoking. In rodents, cigarette smoke and its constituents cause chronic pancreatic inflammation and exocrine damage (43), serving as a precursor of pancreatitis and pancreatic cancer. The contribution of cigarette carcinogens to the development of pancreatic preneoplasms and cancer has been highlighted in vivo, including in EL-IL-1β mice (44) and in a 7,12-dimethylbenz[α]anthracene-induced mouse model, where nicotine provided robust progression of mPanIN to PDAC (45). Identifying the specific molecular and cellular effects of these carcinogens and their accompanying genetic lesions with respect to pancreatic cancer development has not been fully addressed in vivo.

Developmental pathways

Developmental pathways may also promote pancreatic cancer and thereby serve as targets for chemoprevention. Indeed, the co-expression of an active Notch allele (Rosa26NIC; ref. 11) cooperated with Kras-G12D to promote PanIN formation; conversely, the attenuation of the Notch pathway with a gamma secretase inhibitor mitigated mPanIN formation and PDAC formation in Kras; p53fl//+ mice (46). Despite evidence that Notch is oncogenic, conditional loss of Notch in the pancreas of Pdx1-Cre/LSL-Kras mice demonstrated that Notch may act as a tumor suppressor gene (47), which might be related to the context and/or timing of Notch expression. Likewise, TGFβ signals can have a similar dichotomous nature by serving tumor-promoting and -suppressing functions (48, 49), which also may be dependent on context and/or timing. Loss of either smad4 or Tgfbr2 in p48Cre or Pdx1-Cre/LSL-Kras mice led to more aggressive disease (810), yet EL-Kras mice with haploinsufficient Tgfbr1 generated reduced incidence and frequency but greater size of preinvasive lesions (50). Therefore, inhibition of the Notch or TGFβ signaling pathway needs to be approached cautiously with regard to the cell type and disease stage being targeted.

The hedgehog (Hh)-pathway effector Gli2 cooperates with KrasG12D in a cell-autonomous fashion to promote PanIN/PDAC (51), and Hh-pathway inhibitors alone (52) or in combination with gemcitabine (53) were shown to increase survival and decrease metastasis in two models of advanced PDAC. The deletion of Smoothened, however, disrupts the recognition of extracellular Hh ligands without affecting PanIN/PDAC formation in mouse models, demonstrating that Gli signaling is ligand-independent in PanIN/PDAC, with a direct influence on the desmoplastic stroma (54). Terpenoids may also serve to inhibit the Hh (specifically sonic Hh) pathway (55). A more recent approach using triterpenoids and rexinoids alone and in combination demonstrated strong efficacy leading to greatly improved survival in Kras; p53fl//+ mice, as reported elsewhere in this issue of the journal (56). The potential of a similar approach in Kras cohorts with wild-type p53 yielding a potent chemopreventive effect against mPanIN formation would seem reasonable.

Receptor tyrosine kinase (RTK) pathways known to be relevant for normal development have also been implicated as potential targets in PanIN/PDAC, with the epidermal growth factor receptor (EGFR) inhibitor gefitinib causing reduced incidence of mPanINs 1 and 2 as well as suppressing progression to invasive disease in p48Cre/LSL-Kras mice, as reported elsewhere in this issue of the journal (57). The recepteur d’origine nantais (RON) receptor also appears to play several roles, including in motility and invasiveness (58) and vascular endothelial growth factor production in pancreatic cancer cells (59), showing itself to be a target of inhibition prior to cancer dissemination. Recent findings indicate that RON signaling mediates cell survival and gemcitabine resistance in a human pancreatic cancer–derived xenograft system, where short hairpin RNA (shRNA)-induced suppression eventually led to compensatory mechanisms via the c-met and EGFR cascades (60).

Future targets

Potential avenues of pursuit should be based on previous data collected from cell culture and/or xenograft models which demonstrate significant efficacy at various targets. As mentioned above, Hh pathway inhibition altered the stromal composition and increased chemotherapy delivery and response (53). Therefore, individual and combinatorial approaches that alter other components of the pancreatic tumor microenvironment should also be considered as preventive strategies, such as those targeting cancer associated fibroblasts (61), myeloid derived suppressor cells, and T-cells (62).

Food-derived polyphenols, such as quercetin and trans-resveratrol, have exhibited notable pro-apoptotic effects on pancreatic cancer cells in vitro and implanted in nude mice (63). Perillyl alcohol (64) has significant chemopreventive effects on cultured pancreatic cancer cells and, when coupled with adenovirus-mediated mda-7/IL-24 gene therapy in vivo, leads to nearly complete loss of human pancreatic cancer following xenograft implants in nude mice (65, 66). Expression of chemokine (C-X-C motif) ligand 12 (CXCL12) and CXC receptor 4 (CXCR4) is higher in PanIN 3 (compared with normal ducts) in both humans and mice, and a dose-dependent increase in cell proliferation of mPanIN cells was observed in mice treated with CXCL12 (67). Both chemokines were partially dependent on mitogen-activated protein kinase signaling. Indeed, a host of other targets are currently being delineated for potential use in these genetically modified mice (68).

Conclusions

The stage is set to deploy various in vivo models of pancreatic neoplasia for evaluation of multiple chemoprevention strategies against a host of target types including inflammatory, epigenetic, and developmental targets. Both model and target should maintain some aspects of etiology observed in human pancreatic cancer. The challenge is to expand findings from culture and/or xenograft/orthotopic systems into genetically modified models, while preparing to translate these results for future relevance in the clinic (72).

Footnotes

Disclosure of Potential Conflicts of Interest: No potential conflicts of interest were disclosed.

References

  • 1.Brand RE, Lerch MM, Rubinstein WS, Neoptolemos JP, Whitcomb DC, Hruban RH, Brentnall TA, Lynch HT, Canto MI. Advances in counselling and surveillance of patients at risk for pancreatic cancer. Gut. 2007;56:1460–1469. doi: 10.1136/gut.2006.108456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Carter R, Stocken DD, Ghaneh P, Bramhall SR, Olah A, Kelemen D, Bassi C, Friess H, Dervenis C, Spry N, Buchler MW, Neoptolemos JP. Longitudinal quality of life data can provide insights on the impact of adjuvant treatment for pancreatic cancer-Subset analysis of the ESPAC-1 data. Int J Cancer. 2009;124:2960–2965. doi: 10.1002/ijc.24270. [DOI] [PubMed] [Google Scholar]
  • 3.Neoptolemos JP, Stocken DD, Smith C. Tudur, Bassi C, Ghaneh P, Owen E, Moore M, Padbury R, Doi R, Smith D, Buchler MW. Adjuvant 5-fluorouracil and folinic acid vs observation for pancreatic cancer: composite data from the ESPAC-1 and -3(v1) trials. Br J Cancer. 2009;100:246–250. doi: 10.1038/sj.bjc.6604838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ding Y, Cravero JD, Adrian K, Grippo P. Modeling pancreatic cancer in vivo: from xenograft and carcinogen-induced systems to genetically engineered mice. Pancreas. 39:283–292. doi: 10.1097/MPA.0b013e3181c15619. [DOI] [PubMed] [Google Scholar]
  • 5.Hruban RH, Adsay NV, Albores-Saavedra J, Anver MR, Biankin AV, Boivin GP, Furth EE, Furukawa T, Klein A, Klimstra DS, Kloppel G, Lauwers GY, Longnecker DS, Luttges J, Maitra A, Offerhaus GJ, Perez-Gallego L, Redston M, Tuveson DA. Pathology of genetically engineered mouse models of pancreatic exocrine cancer: consensus report and recommendations. Cancer Res. 2006;66:95–106. doi: 10.1158/0008-5472.CAN-05-2168. [DOI] [PubMed] [Google Scholar]
  • 6.Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, Redston MS, DePinho RA. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev. 2003;17:3112–3126. doi: 10.1101/gad.1158703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469–483. doi: 10.1016/j.ccr.2005.04.023. [DOI] [PubMed] [Google Scholar]
  • 8.Izeradjene K, Combs C, Best M, Gopinathan A, Wagner A, Grady WM, Deng CX, Hruban RH, Adsay NV, Tuveson DA, Hingorani SR. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell. 2007;11:229–243. doi: 10.1016/j.ccr.2007.01.017. [DOI] [PubMed] [Google Scholar]
  • 9.Kojima K, Vickers SM, Adsay NV, Jhala NC, Kim HG, Schoeb TR, Grizzle WE, Klug CA. Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Res. 2007;67:8121–8130. doi: 10.1158/0008-5472.CAN-06-4167. [DOI] [PubMed] [Google Scholar]
  • 10.Ijichi H, Chytil A, Gorska AE, Aakre ME, Fujitani Y, Fujitani S, Wright CV, Moses HL. Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes Dev. 2006;20:3147–3160. doi: 10.1101/gad.1475506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.De La OJ, Emerson LL, Goodman JL, Froebe SC, Illum BE, Curtis AB, Murtaugh LC. Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia. Proc Natl Acad Sci U S A. 2008;105:18907–18912. doi: 10.1073/pnas.0810111105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gidekel Friedlander SY, Chu GC, Snyder EL, Girnius N, Dibelius G, Crowley D, Vasile E, DePinho RA, Jacks T. Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell. 2009;16:379–389. doi: 10.1016/j.ccr.2009.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291–302. doi: 10.1016/j.ccr.2007.01.012. [DOI] [PubMed] [Google Scholar]
  • 14.Habbe N, Shi G, Meguid RA, Fendrich V, Esni F, Chen H, Feldmann G, Stoffers DA, Konieczny SF, Leach SD, Maitra A. Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice. Proc Natl Acad Sci U S A. 2008;105:18913–18918. doi: 10.1073/pnas.0810097105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ji B, Tsou L, Wang H, Gaiser S, Chang DZ, Daniluk J, Bi Y, Grote T, Longnecker DS, Logsdon CD. Ras activity levels control the development of pancreatic diseases. Gastroenterology. 2009;137:1072–1082. 1082, e1071–1076. doi: 10.1053/j.gastro.2009.05.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hingorani SR. A New Preclinical Paradigm for Pancreas Cancer. Springer; 2010. pp. 73–93. [Google Scholar]
  • 17.Olive KP, Tuveson DA. The use of targeted mouse models for preclinical testing of novel cancer therapeutics. Clin Cancer Res. 2006;12:5277–5287. doi: 10.1158/1078-0432.CCR-06-0436. [DOI] [PubMed] [Google Scholar]
  • 18.Whitcomb DC, Gorry MC, Preston RA, Furey W, Sossenheimer MJ, Ulrich CD, Martin SP, Gates LK, Jr., Amann ST, Toskes PP, Liddle R, McGrath K, Uomo G, Post JC, Ehrlich GD. Hereditary pancreatitis is caused by a mutation in the cationic trypsinogen gene. Nat Genet. 1996;14:141–145. doi: 10.1038/ng1096-141. [DOI] [PubMed] [Google Scholar]
  • 19.Archer H, Jura N, Keller J, Jacobson M, Bar-Sagi D. A mouse model of hereditary pancreatitis generated by transgenic expression of R122H trypsinogen. Gastroenterology. 2006;131:1844–1855. doi: 10.1053/j.gastro.2006.09.049. [DOI] [PubMed] [Google Scholar]
  • 20.Carriere C, Young AL, Gunn JR, Longnecker DS, Korc M. Acute pancreatitis markedly accelerates pancreatic cancer progression in mice expressing oncogenic Kras. Biochem Biophys Res Commun. 2009;382:561–565. doi: 10.1016/j.bbrc.2009.03.068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Selig L, Sack U, Gaiser S, Kloppel G, Savkovic V, Mossner J, Keim V, Bodeker H. Characterisation of a transgenic mouse expressing R122H human cationic trypsinogen. BMC Gastroenterol. 2006;6:30. doi: 10.1186/1471-230X-6-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, Kawaguchi Y, Johann D, Liotta LA, Crawford HC, Putt ME, Jacks T, Wright CV, Hruban RH, Lowy AM, Tuveson DA. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4:437–450. doi: 10.1016/s1535-6108(03)00309-x. [DOI] [PubMed] [Google Scholar]
  • 23.Funahashi H, Satake M, Dawson D, Huynh NA, Reber HA, Hines OJ, Eibl G. Delayed progression of pancreatic intraepithelial neoplasia in a conditional Kras(G12D) mouse model by a selective cyclooxygenase-2 inhibitor. Cancer Res. 2007;67:7068–7071. doi: 10.1158/0008-5472.CAN-07-0970. [DOI] [PubMed] [Google Scholar]
  • 24.Colby JK, Klein RD, McArthur MJ, Conti CJ, Kiguchi K, Kawamoto T, Riggs PK, Pavone AI, Sawicki J, Fischer SM. Progressive metaplastic and dysplastic changes in mouse pancreas induced by cyclooxygenase-2 overexpression. Neoplasia. 2008;10:782–796. doi: 10.1593/neo.08330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mukherjee P, Basu GD, Tinder TL, Subramani DB, Bradley JM, Arefayene M, Skaar T, De Petris G. Progression of pancreatic adenocarcinoma is significantly impeded with a combination of vaccine and COX-2 inhibition. J Immunol. 2009;182:216–224. [PMC free article] [PubMed] [Google Scholar]
  • 26.Ishikawa TO, Herschman HR. Conditional knockout mouse for tissue-specific disruption of the cyclooxygenase-2 (Cox-2) gene. Genesis. 2006;44:143–149. doi: 10.1002/gene.20192. [DOI] [PubMed] [Google Scholar]
  • 27.Fujioka S, Sclabas GM, Schmidt C, Frederick WA, Dong QG, Abbruzzese JL, Evans DB, Baker C, Chiao PJ. Function of nuclear factor kappaB in pancreatic cancer metastasis. Clin Cancer Res. 2003;9:346–354. [PubMed] [Google Scholar]
  • 28.Sclabas GM, Uwagawa T., Schmidt, C., Hess KR, Evans DB, Abbruzzese JL, Chiao PJ. Nuclear factor kappa B activation is a potential target for preventing pancreatic carcinoma by aspirin. Cancer. 2005;103:2485–2490. doi: 10.1002/cncr.21075. [DOI] [PubMed] [Google Scholar]
  • 29.Zhang Z, Rigas B. NF-kappaB, inflammation and pancreatic carcinogenesis: NF-kappaB as a chemoprevention target (review) Int J Oncol. 2006;29:185–192. [PubMed] [Google Scholar]
  • 30.Hennig R, Grippo P, Ding XZ, Rao SM, Buchler MW, Friess H, Talamonti MS, Bell RH, Adrian TE. 5-Lipoxygenase, a marker for early pancreatic intraepithelial neoplastic lesions. Cancer Res. 2005;65:6011–6016. doi: 10.1158/0008-5472.CAN-04-4090. [DOI] [PubMed] [Google Scholar]
  • 31.Tong WG, Ding XZ, Hennig R, Witt RC, Standop J, Pour PM, Adrian TE. Leukotriene B4 receptor antagonist LY293111 inhibits proliferation and induces apoptosis in human pancreatic cancer cells. Clin Cancer Res. 2002;8:3232–3242. [PubMed] [Google Scholar]
  • 32.Tong WG, Ding XZ, Talamonti MS, Bell RH, Adrian TE. Leukotriene B4 receptor antagonist LY293111 induces S-phase cell cycle arrest and apoptosis in human pancreatic cancer cells. Anticancer Drugs. 2007;18:535–541. doi: 10.1097/01.cad.0000231477.22901.8a. [DOI] [PubMed] [Google Scholar]
  • 33.Hennig R, Ventura J, Segersvard R, Ward E, Ding XZ, Rao SM, Jovanovic BD, Iwamura T, Talamonti MS, Bell RH, Jr., Adrian TE. LY293111 improves efficacy of gemcitabine therapy on pancreatic cancer in a fluorescent orthotopic model in athymic mice. Neoplasia. 2005;7:417–425. doi: 10.1593/neo.04559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Zyromski NJ, Mathur A, Gowda GA, Murphy C, Swartz-Basile DA, Wade TE, Pitt HA, Raftery D. Nuclear magnetic resonance spectroscopy-based metabolomics of the fatty pancreas: implicating fat in pancreatic pathology. Pancreatology. 2009;9:410–419. doi: 10.1159/000199436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zyromski NJ, Mathur A, Pitt HA, Wade TE, Wang S, Nakshatri P, Swartz-Basile DA, Nakshatri H. Obesity potentiates the growth and dissemination of pancreatic cancer. Surgery. 2009;146:258–263. doi: 10.1016/j.surg.2009.02.024. [DOI] [PubMed] [Google Scholar]
  • 36.Khasawneh J, Schulz MD, Walch A, Rozman J, Hrabe de Angelis M, Klingenspor M, Buck A, Schwaiger M, Saur D, Schmid RM, Kloppel G, Sipos B, Greten FR, Arkan MC. Inflammation and mitochondrial fatty acid beta-oxidation link obesity to early tumor promotion. Proc Natl Acad Sci U S A. 2009;106:3354–3359. doi: 10.1073/pnas.0802864106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Strouch MJ, Ding Y, Salabat MR, Melstrom LG, Adrian K, Quinn C, Pelham C, Rao S, Adrian TE, Bentrem DJ, Grippo PJ. A High Omega-3 Fatty Acid Diet Mitigates Murine Pancreatic Precancer Development. J Surg Res. 2009 doi: 10.1016/j.jss.2009.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liu H, Huang D, McArthur DL, Boros LG, Nissen N, Heaney AP. Fructose induces transketolase flux to promote pancreatic cancer growth. Cancer Res. 70:6368–6376. doi: 10.1158/0008-5472.CAN-09-4615. [DOI] [PubMed] [Google Scholar]
  • 39.Mueller NT, Odegaard A, Anderson K, Yuan JM, Gross M, Koh WP, Pereira MA. Soft drink and juice consumption and risk of pancreatic cancer: the Singapore Chinese Health Study. Cancer Epidemiol Biomarkers Prev. 19:447–455. doi: 10.1158/1055-9965.EPI-09-0862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rossi M, Lipworth L, Polesel J, Negri E, Bosetti C, Talamini R, McLaughlin JK, La Vecchia C. Dietary glycemic index and glycemic load and risk of pancreatic cancer: a case-control study. Ann Epidemiol. 20:460–465. doi: 10.1016/j.annepidem.2010.03.018. [DOI] [PubMed] [Google Scholar]
  • 41.Simon MS, Shikany JM, Neuhouser ML, Rohan T, Nirmal K, Cui Y, Abrams J. Glycemic index, glycemic load, and the risk of pancreatic cancer among postmenopausal women in the women’s health initiative observational study and clinical trial. Cancer Causes Control. doi: 10.1007/s10552-010-9632-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pannala R, Basu A, Petersen GM, Chari ST. New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol. 2009;10:88–95. doi: 10.1016/S1470-2045(08)70337-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wittel UA, Hopt UT, Batra SK. Cigarette smoke-induced pancreatic damage: experimental data. Langenbecks Arch Surg. 2008;393:581–588. doi: 10.1007/s00423-007-0273-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Song Z, Bhagat G, Quante M, Baik GH, Marrache F, Tu SP, Zhao CM, Chen D, Dannenberg AJ, Wang TC. Potential carcinogenic effects of cigarette smoke and Swedish moist snuff on pancreas: a study using a transgenic mouse model of chronic pancreatitis. Lab Invest. 90:426–435. doi: 10.1038/labinvest.2009.145. [DOI] [PubMed] [Google Scholar]
  • 45.Bersch VP, Osvaldt AB, Edelweiss MI, Schumacher Rde C, Wendt LR, Abreu LP, Blom CB, Abreu GP, Costa L, Piccinini P, Rohde L. Effect of nicotine and cigarette smoke on an experimental model of intraepithelial lesions and pancreatic adenocarcinoma induced by 7,12-dimethylbenzanthracene in mice. Pancreas. 2009;38:65–70. doi: 10.1097/MPA.0b013e318184d330. [DOI] [PubMed] [Google Scholar]
  • 46.Plentz R, Park JS, Rhim AD, Abravanel D, Hezel AF, Sharma SV, Gurumurthy S, Deshpande V, Kenific C, Settleman J, Majumder PK, Stanger BZ, Bardeesy N. Inhibition of gamma-secretase activity inhibits tumor progression in a mouse model of pancreatic ductal adenocarcinoma. Gastroenterology. 2009;136:1741–1749. e1746. doi: 10.1053/j.gastro.2009.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hanlon L, Avila JL, Demarest RM, Troutman S, Allen M, Ratti F, Rustgi AK, Stanger BZ, Radtke F, Adsay V, Long F, Capobianco AJ, Kissil JL. Notch1 functions as a tumor suppressor in a model of K-ras-induced pancreatic ductal adenocarcinoma. Cancer Res. 70:4280–4286. doi: 10.1158/0008-5472.CAN-09-4645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Derynck R, Akhurst RJ, Balmain A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001;29:117–129. doi: 10.1038/ng1001-117. [DOI] [PubMed] [Google Scholar]
  • 49.Schniewind B, Groth S, Sebens Muerkoster S, Sipos B, Schafer H, Kalthoff H, Fandrich F, Ungefroren H. Dissecting the role of TGF-beta type I receptor/ALK5 in pancreatic ductal adenocarcinoma: Smad activation is crucial for both the tumor suppressive and prometastatic function. Oncogene. 2007;26:4850–4862. doi: 10.1038/sj.onc.1210272. [DOI] [PubMed] [Google Scholar]
  • 50.Adrian K, Strouch MJ, Zeng Q, Barron MR, Cheon EC, Honasoge A, Xu Y, Phukan S, Sadim M, Bentrem DJ, Pasche B, Grippo PJ. Tgfbr1 haploinsufficiency inhibits the development of murine mutant Kras-induced pancreatic precancer. Cancer Res. 2009;69:9169–9174. doi: 10.1158/0008-5472.CAN-09-1705. [DOI] [PubMed] [Google Scholar]
  • 51.Pasca di Magliano M, Sekine S, Ermilov A, Ferris J, Dlugosz AA, Hebrok M. Hedgehog/Ras interactions regulate early stages of pancreatic cancer. Genes Dev. 2006;20:3161–3173. doi: 10.1101/gad.1470806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Feldmann G, Habbe N, Dhara S, Bisht S, Alvarez H, Fendrich V, Beaty R, Mullendore M, Karikari C, Bardeesy N, Ouellette MM, Yu W, Maitra A. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut. 2008;57:1420–1430. doi: 10.1136/gut.2007.148189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D, Frese KK, Denicola G, Feig C, Combs C, Winter SP, Ireland-Zecchini H, Reichelt S, Howat WJ, Chang A, Dhara M, Wang L, Ruckert F, Grutzmann R, Pilarsky C, Izeradjene K, Hingorani SR, Huang P, Davies SE, Plunkett W, Egorin M, Hruban RH, Whitebread N, McGovern K, Adams J, Iacobuzio-Donahue C, Griffiths J, Tuveson DA. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457–1461. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nolan-Stevaux O, Lau J, Truitt ML, Chu GC, Hebrok M, Fernandez-Zapico ME, Hanahan D. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev. 2009;23:24–36. doi: 10.1101/gad.1753809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rifai Y, Arai MA, Koyano T, Kowithayakorn T, Ishibashi M. Terpenoids and a flavonoid glycoside from Acacia pennata leaves as hedgehog/GLI-mediated transcriptional inhibitors. J Nat Prod. 73:995–997. doi: 10.1021/np1000818. [DOI] [PubMed] [Google Scholar]
  • 56.Liby K, R. D, Risingsong R, et al. Synthetic triterpenoids prolong survival in a transgenic mouse model of pancreatic cancer. Cancer Prev Res. 2010;3:1427–1234. doi: 10.1158/1940-6207.CAPR-10-0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mohammed A, J. N, Li Q, et al. EGFR inhibitor gefitnib prevents progression of pancreatic lesions to carcinoma in a conditional LSL-KrasG12D/+ transgenic mouse model. Cancer Prev Res. 2010;3:1417–1426. doi: 10.1158/1940-6207.CAPR-10-0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhao S, Ammanamanchi S, Brattain M, Cao L, Thangasamy A, Wang J, Freeman JW. Smad4-dependent TGF-beta signaling suppresses RON receptor tyrosine kinase-dependent motility and invasion of pancreatic cancer cells. J Biol Chem. 2008;283:11293–11301. doi: 10.1074/jbc.M800154200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Thomas RM, Jaquish DV, French RP, Lowy AM. The RON tyrosine kinase receptor regulates vascular endothelial growth factor production in pancreatic cancer cells. Pancreas. 39:301–307. doi: 10.1097/mpa.0b013e3181bb9f73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, Waltz SE, Lowy AM. Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res. 70:1130–1140. doi: 10.1158/0008-5472.CAN-09-0761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell. 17:135–147. doi: 10.1016/j.ccr.2009.12.041. [DOI] [PubMed] [Google Scholar]
  • 62.Tseng WW, Winer D, Kenkel JA, Choi O, Shain AH, Pollack JR, French R, Lowy AM, Engleman EG. Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clin Cancer Res. 16:3684–3695. doi: 10.1158/1078-0432.CCR-09-2384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mouria M, Gukovskaya AS, Jung Y, Buechler P, Hines OJ, Reber HA, Pandol SJ. Food-derived polyphenols inhibit pancreatic cancer growth through mitochondrial cytochrome C release and apoptosis. Int J Cancer. 2002;98:761–769. doi: 10.1002/ijc.10202. [DOI] [PubMed] [Google Scholar]
  • 64.Muller-Decker K, Furstenberger G, Annan N, Kucher D, Pohl-Arnold A, Steinbauer B, Esposito I, Chiblak S, Friess H, Schirmacher P, Berger I. Preinvasive duct-derived neoplasms in pancreas of keratin 5-promoter cyclooxygenase-2 transgenic mice. Gastroenterology. 2006;130:2165–2178. doi: 10.1053/j.gastro.2006.03.053. [DOI] [PubMed] [Google Scholar]
  • 65.Burke YD, Ayoubi AS, Werner SR, McFarland BC, Heilman DK, Ruggeri BA, Crowell PL. Effects of the isoprenoids perillyl alcohol and farnesol on apoptosis biomarkers in pancreatic cancer chemoprevention. Anticancer Res. 2002;22:3127–3134. [PubMed] [Google Scholar]
  • 66.Lebedeva IV, Su ZZ, Vozhilla N, Chatman L, Sarkar D, Dent P, Athar M, Fisher PB. Chemoprevention by perillyl alcohol coupled with viral gene therapy reduces pancreatic cancer pathogenesis. Mol Cancer Ther. 2008;7:2042–2050. doi: 10.1158/1535-7163.MCT-08-0245. [DOI] [PubMed] [Google Scholar]
  • 67.Thomas RM, Kim J, Revelo-Penafiel MP, Angel R, Dawson DW, Lowy AM. The chemokine receptor CXCR4 is expressed in pancreatic intraepithelial neoplasia. Gut. 2008;57:1555–1560. doi: 10.1136/gut.2007.143941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Stan SD, Singh SV, Brand RE. Chemoprevention strategies for pancreatic cancer. Nat Rev Gastroenterol Hepatol. 7:347–356. doi: 10.1038/nrgastro.2010.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Grippo PJ, Nowlin PS, Demeure MJ, Longnecker DS, Sandgren EP. Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Res. 2003;63:2016–2019. [PubMed] [Google Scholar]
  • 70.Tuveson DA, Zhu L, Gopinathan A, Willis NA, Kachatrian L, Grochow R, Pin CL, Mitin NY, Taparowsky EJ, Gimotty PA, Hruban RH, Jacks T, Konieczny SF. Mist1-KrasG12D knock-in mice develop mixed differentiation metastatic exocrine pancreatic carcinoma and hepatocellular carcinoma. Cancer Res. 2006;66:242–247. doi: 10.1158/0008-5472.CAN-05-2305. [DOI] [PubMed] [Google Scholar]
  • 71.Carriere C, Seeley ES, Goetze T, Longnecker DS, Korc M. The Nestin progenitor lineage is the compartment of origin for pancreatic intraepithelial neoplasia. Proc Natl Acad Sci U S A. 104:4437–4442. doi: 10.1073/pnas.0701117104. 2007.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Logsdon CD, Abbruzzese JL. Chemoprevention of pancreatic cancer: ready for the clinic? Cancer Prev Res. 2010;3:1375–1378. doi: 10.1158/1940-6207.CAPR-10-0216. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES