Gastroenterology

Gastroenterology

Volume 142, Issue 5, May 2012, Pages 1079-1092
Gastroenterology

Reviews and Perspectives
Reviews in Basic and Clinical Gastroenterology and Hepatology
What We Have Learned About Pancreatic Cancer From Mouse Models

https://doi.org/10.1053/j.gastro.2012.03.002Get rights and content

Section snippets

Conditional Kras Models of Pancreatic Cancer

Although approaches to generate mouse models of pancreatic cancer started in the late 1980s,2, 3 the GEMM that most closely resembles human disease was established in 2003.4 This model is based on the LSL-KrasG12D strain of mice, which has an endogenous, conditional KrasG12D mutant allele silenced by a floxed transcriptional STOP cassette (Lox-Stop-Lox or LSL) inserted upstream of the targeted Exon1. Removal of this LSL by directing expression of Cre recombinase with adenoviral-Cre allows

Cancer-Initiating Cells in PDA

The ductal-like properties of cells in PanIN lesions indicated that PDAs might derive from normal ductal cells or their precursors.6, 7 However, attempts to induce PanINs from ductal cells, by regulating expression of Kras oncogenes with the cytokeratin-19 promoter, did not generate PanINs or PDA.26 These observations raised doubts that PanINs, despite their ductal-like properties, arise from the ductal cell lineage. In fact, the development of another GEMM similar to the original strain

Pancreatitis and PDA

Adult mice that express endogenous Kras oncogenes in the acinar cell compartment develop PanINs and PDAs with high penetrance when subjected to acute or chronic cerulein-induced pancreatitis. Sporadic bouts of pancreatitis that last as little as 1 month are sufficient to induce PDAs in these mice (Figure 1). Longer bouts (3 months) or chronic pancreatitis increase the tumor incidence and reduce latency.27, 36 Expression of KrasG12V does not have to precede or coincide with pancreatitis,

Inflammation and PDA Development

GEMMs have been used to study how the inflammatory response contributes to the progression of PanINs to yield PDA. In a recent study, Guerra et al showed that the inflammatory response inhibited senescence in low-grade PanINs.36 Interestingly, expression of oncogenic Kras did not induce senescence in normal pancreatic cells; markers of senescence could be detected only in Kras oncogene-expressing cells that were undergoing the transition from acinar to ductal metaplasia or in low-grade PanINs.36

The Ras Effector Pathway in PDA

Activating mutations at codons 12, 13, and 61 in the Kras gene are present in ∼90% of PDAs.49 Oncogenic forms of Kras show impaired intrinsic and extrinsic guanosine triphosphatase function and thereby remain bound to guanosine triphosphate to enable constitutive interactions with downstream effectors (more than 20 are known) that regulate cell responses such as proliferation migration, differentiation, adhesion, survival, and apoptosis.50, 51 The most well-studied Ras effectors include

Using Mouse Models to Identify and Validate Human PDA Genes

During the past decade, many new genes and biochemical pathways have been associated with the progression of pancreatic cancer. The whole exome sequencing and copy number analyses of 24 human tumors have identified more than 1000 somatic mutations, with an average of 63 per tumor.86 These mutant alleles have been assigned to 12 proposed core pathways of PDA.86 Determining the role of such genes in PDA initiation and progression is important and could uncover new therapeutic approaches.

Development of Diagnostic and Therapeutic Strategies for PDA

GEMMs might be used to identify markers of early-stage pancreatic cancer because no good markers are currently available for early detection of patients. The PanIN models are ideal for development of tissue biomarkers of pancreatic preneoplasia; a low-resolution serum proteomic analyses provided initial evidence that such an approach could succeed.4 A more quantitative approach revealed multiple potential plasma biomarkers of pancreatic preneoplasia in GEMMs.99 Additional work by Kelly et al

Conclusions

Over the past decade, GEMMs of PDA have increased our understanding of this disease and allowed initial exploration of therapeutic and diagnostic strategies. Refinements of these first-generation models will continue to provide new insights into the genes, pathways, and cell types involved in the pathogenesis of PDA. Nonetheless, these freely available models should be expeditiously used by our community to identify better diagnostic and therapeutic options for patients. GEMMs could also be

Acknowledgments

The authors thank Dr A. Neesse and Dr F. Connor for critically reviewing the manuscript; Dr C. Feig for providing images of mouse pancreatitis; Dr N. Cook, Dr K. Freese, and Dr D. Lewis for providing the FDG-PET/CT image; and Paul Mackin for providing the high-resolution ultrasonography image.

First page preview

First page preview
Click to open first page preview

References (138)

  • C. Guerra et al.

    Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence

    Cancer Cell

    (2011)
  • P.M. Rothwell et al.

    Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials

    Lancet

    (2011)
  • K.E. Lee et al.

    Oncogenic KRas suppresses inflammation-associated senescence of pancreatic ductal cells

    Cancer Cell

    (2010)
  • A. Fukuda et al.

    Stat3 and MMP7 contribute to pancreatic ductal adenocarcinoma initiation and progression

    Cancer Cell

    (2011)
  • M. Lesina et al.

    Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer

    Cancer Cell

    (2011)
  • K. Rajalingam et al.

    Ras oncogenes and their downstream targets

    Biochim Biophys Acta

    (2007)
  • N. Mitin et al.

    Signaling interplay in Ras superfamily function

    Curr Biol

    (2005)
  • M.T. Yip-Schneider et al.

    Pancreatic tumor cells with mutant K-ras suppress ERK activity by MEK-dependent induction of MAP kinase phosphatase-2

    Biochem Biophys Res Commun

    (2001)
  • E.S. Calhoun et al.

    BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets

    Am J Pathol

    (2003)
  • R.B. Blasco et al.

    c-Raf, but not B-Raf, is essential for development of K-Ras oncogene-driven non-small cell lung carcinoma

    Cancer Cell

    (2011)
  • M.A. White et al.

    Multiple Ras functions can contribute to mammalian cell transformation

    Cell

    (1995)
  • P. Rodriguez-Viciana et al.

    Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras

    Cell

    (1997)
  • J. Luo et al.

    Targeting the PI3K-Akt pathway in human cancer: rationale and promise

    Cancer Cell

    (2003)
  • A.L. Kennedy et al.

    Activation of the PIK3CA/AKT pathway suppresses senescence induced by an activated RAS oncogene to promote tumorigenesis

    Mol Cell

    (2011)
  • A. Rangarajan et al.

    Species- and cell type-specific requirements for cellular transformation

    Cancer Cell

    (2004)
  • K.H. Lim et al.

    Activation of RalA is critical for Ras-induced tumorigenesis of human cells

    Cancer Cell

    (2005)
  • K.H. Lim et al.

    Divergent roles for RalA and RalB in malignant growth of human pancreatic carcinoma cells

    Curr Biol

    (2006)
  • J. Jonkers et al.

    Retroviral insertional mutagenesis as a strategy to identify cancer genes

    Biochim Biophys Acta

    (1996)
  • Z. Ivics et al.

    Molecular reconstruction of Sleeping Beauty, a Tc1-like transposon from fish, and its transposition in human cells

    Cell

    (1997)
  • S. Ding et al.

    Efficient transposition of the piggyBac (PB) transposon in mammalian cells and mice

    Cell

    (2005)
  • A. Jemal et al.

    Cancer statistics, 2010

    CA Cancer J Clin

    (2010)
  • D.M. Ornitz et al.

    Pancreatic neoplasia induced by SV40 T-antigen expression in acinar cells of transgenic mice

    Science

    (1987)
  • E.L. Jackson et al.

    Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras

    Genes Dev

    (2001)
  • R.H. Hruban et al.

    Progression model for pancreatic cancer

    Clin Cancer Res

    (2000)
  • C. Caldas et al.

    Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma

    Nat Genet

    (1994)
  • M.S. Redston et al.

    p53 mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions

    Cancer Res

    (1994)
  • S.A. Hahn et al.

    DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1

    Science

    (1996)
  • M. Goggins et al.

    Genetic alterations of the transforming growth factor beta receptor genes in pancreatic and biliary adenocarcinomas

    Cancer Res

    (1998)
  • A.J. Aguirre et al.

    Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma

    Genes Dev

    (2003)
  • N. Bardeesy et al.

    Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse

    Proc Natl Acad Sci U S A

    (2006)
  • H. Ijichi et al.

    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)
  • E.A. Collisson et al.

    Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy

    Nat Med

    (2011)
  • A. Maitra et al.

    Precursors to invasive pancreatic cancer

    Adv Anat Pathol

    (2005)
  • N. Bardeesy et al.

    Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer

    Genes Dev

    (2006)
  • D.F. Vincent et al.

    Inactivation of TIF1gamma cooperates with Kras to induce cystic tumors of the pancreas

    PLoS Genet

    (2009)
  • P.J. Grippo et al.

    Deploying mouse models of pancreatic cancer for chemoprevention studies

    Cancer Prev Res (Phila)

    (2010)
  • P.K. Mazur et al.

    Genetically engineered mouse models of pancreatic cancer: unravelling tumour biology and progressing translational oncology

    Gut

    (2011 Aug 26)
  • A. Neesse et al.

    Stromal biology and therapy in pancreatic cancer

    Gut

    (2011)
  • F.H. Brembeck et al.

    The mutant K-ras oncogene causes pancreatic periductal lymphocytic infiltration and gastric mucous neck cell hyperplasia in transgenic mice

    Cancer Res

    (2003)
  • N. Habbe et al.

    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)
  • Cited by (146)

    • Ductal metaplasia in pancreas

      2022, Biochimica et Biophysica Acta - Reviews on Cancer
    • KRAS mutation in pancreatic cancer

      2021, Seminars in Oncology
    View all citing articles on Scopus

    Conflicts of interest The authors disclose no conflicts.

    View full text