Genetically engineered mouse models of pancreatic cancer.

Pancreatic ductal adenocarcinoma is the 10th most common cancer and the fourth leading cause of cancer-related death in the United States. Despite great effort, the prognosis for patients with this disease remains dismal with a 5-year survival rate of just 4% to 6%. Although several important advances have improved our understanding of the underlying biology of pancreatic cancer, this knowledge has not translated into novel therapeutic approaches and effective systemic or targeted therapies. Pancreatic cancer is one of the malignancies most difficult to treat, with remarkable intrinsic resistance to both standard and targeted chemotherapy as well as ionizing radiation. Surgical intervention remains the only potentially curative approach. However, most patients present with inoperable and/or metastatic disease and are therefore excluded from surgery. Accordingly, new therapeutic options are desperately needed. In vivo models to study innovative and alternative treatment approaches are of major importance. A variety of genetically engineered mouse models of pancreatic cancer have been developed over the last decade. However, these models display different characteristics, and not all of them are suited for preclinical studies. In this review, we aim to review the mouse models available, their experimental use, their clinical relevance and limitations, and future directions.

[1]  C. Der,et al.  Biological and biochemical properties of human ras H genes mutated at codon 61 , 1986, Cell.

[2]  E. Petricoin,et al.  Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. , 2003, Cancer cell.

[3]  T. Skaar,et al.  MUC1 Enhances Tumor Progression and Contributes Toward Immunosuppression in a Mouse Model of Spontaneous Pancreatic Adenocarcinoma1 , 2008, The Journal of Immunology.

[4]  S. Dry,et al.  PTEN loss accelerates KrasG12D-induced pancreatic cancer development. , 2010, Cancer research.

[5]  I. Bross,et al.  Environmental factors in cancer of the upper alimentary tract. A swedish study with special reference to plummer‐vinson (Paterson‐Kelly) syndrome , 1957, Cancer.

[6]  J B Gibbs,et al.  Intrinsic GTPase activity distinguishes normal and oncogenic ras p21 molecules. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[7]  F. Schabel,et al.  Induction and chemotherapeutic response of two transplantable ductal adenocarcinomas of the pancreas in C57BL/6 mice. , 1984, Cancer research.

[8]  D. Saur,et al.  RCAS-mediated retroviral gene delivery: a versatile tool for the study of gene function in a mouse model of pancreatic cancer. , 2008, Human gene therapy.

[9]  Changes in Connexin43 Expression and Localization During Pancreatic Cancer Progression , 2012, The Journal of Membrane Biology.

[10]  Drew A. Torigian,et al.  CD40 Agonists Alter Tumor Stroma and Show Efficacy Against Pancreatic Carcinoma in Mice and Humans , 2011, Science.

[11]  P. Spellman,et al.  Subtypes of Pancreatic Ductal Adenocarcinoma and Their Differing Responses to Therapy , 2011, Nature Medicine.

[12]  Fergus J Couch,et al.  Inactivation of Brca2 promotes Trp53-associated but inhibits KrasG12D-dependent pancreatic cancer development in mice. , 2011, Gastroenterology.

[13]  C. Wright,et al.  Ongoing Notch signaling maintains phenotypic fidelity in the adult exocrine pancreas. , 2012, Developmental biology.

[14]  T. Wang,et al.  Stem cells in gastroenterology and hepatology , 2009, Nature Reviews Gastroenterology &Hepatology.

[15]  Thomas G. Caffery,et al.  Sonic Hedgehog Promotes Desmoplasia in Pancreatic Cancer , 2008, Clinical Cancer Research.

[16]  D. Tuveson,et al.  Crosstalk between the canonical NF-κB and Notch signaling pathways inhibits Pparγ expression and promotes pancreatic cancer progression in mice. , 2011, The Journal of clinical investigation.

[17]  Derek S. Chan,et al.  Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer , 2012, Gut.

[18]  R. Schmid,et al.  Concomitant pancreatic activation of Kras(G12D) and Tgfa results in cystic papillary neoplasms reminiscent of human IPMN. , 2007, Cancer cell.

[19]  R. DePinho,et al.  Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. , 2003, Genes & development.

[20]  A. Capobianco,et al.  Notch signalling in solid tumours: a little bit of everything but not all the time , 2011, Nature Reviews Cancer.

[21]  H. Friess,et al.  From tissue turnover to the cell of origin for pancreatic cancer , 2011, Nature Reviews Gastroenterology &Hepatology.

[22]  A. Ashworth,et al.  LKB1 Haploinsufficiency Cooperates With Kras to Promote Pancreatic Cancer Through Suppression of p21-Dependent Growth Arrest , 2010, Gastroenterology.

[23]  D. Melton,et al.  Direct evidence for the pancreatic lineage: NGN3+ cells are islet progenitors and are distinct from duct progenitors. , 2002, Development.

[24]  T. Mattfeldt,et al.  A murine tumor progression model for pancreatic cancer recapitulating the genetic alterations of the human disease. , 2001, Genes & development.

[25]  J. L. Goodman,et al.  Notch and Kras reprogram pancreatic acinar cells to ductal intraepithelial neoplasia , 2008, Proceedings of the National Academy of Sciences.

[26]  P. Mazur,et al.  Early requirement of Rac1 in a mouse model of pancreatic cancer. , 2011, Gastroenterology.

[27]  R. DePinho,et al.  Pten constrains centroacinar cell expansion and malignant transformation in the pancreas. , 2005, Cancer cell.

[28]  H. Tian,et al.  Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis , 2009, Proceedings of the National Academy of Sciences.

[29]  C. Galbán,et al.  Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. , 2012, The Journal of clinical investigation.

[30]  H. Moses,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. , 2006, Genes & development.

[31]  D. Goeddel,et al.  Comparative biochemical properties of normal and activated human ras p21 protein , 1984, Nature.

[32]  M. Sander,et al.  Sox9+ ductal cells are multipotent progenitors throughout development but do not produce new endocrine cells in the normal or injured adult pancreas , 2011, Development.

[33]  M. Mullendore,et al.  An orally bioavailable small-molecule inhibitor of Hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer , 2008, Molecular Cancer Therapeutics.

[34]  Frank Buchholz,et al.  Dre recombinase, like Cre, is a highly efficient site-specific recombinase in E. coli, mammalian cells and mice , 2009, Disease Models & Mechanisms.

[35]  Gerald C. Chu,et al.  Pancreatic Lkb1 Deletion Leads to Acinar Polarity Defects and Cystic Neoplasms , 2008, Molecular and Cellular Biology.

[36]  M. Hebrok,et al.  Beta-catenin blocks Kras-dependent reprogramming of acini into pancreatic cancer precursor lesions in mice. , 2010, The Journal of clinical investigation.

[37]  N. Jhala,et al.  Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. , 2012, Cancer cell.

[38]  B. Bao,et al.  Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. , 2011, Cancer letters.

[39]  C. Wright,et al.  The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors , 2002, Nature Genetics.

[40]  Gregory Y. Lauwers,et al.  Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis , 2003, Nature.

[41]  D. Longnecker Lesions induced in rodent pancreas by azaserine and other pancreatic carcinogens. , 1984, Environmental health perspectives.

[42]  H. Kocher,et al.  Pancreatic Cancer , 2019, Methods in Molecular Biology.

[43]  T. Jacks,et al.  Somatic activation of the K-ras oncogene causes early onset lung cancer in mice , 2001, Nature.

[44]  L. Chin,et al.  Non-germline genetically engineered mouse models for translational cancer research , 2010, Nature Reviews Cancer.

[45]  D. Saur,et al.  A Cre-loxP-based mouse model for conditional somatic gene expression and knockdown in vivo by using avian retroviral vectors , 2008, Proceedings of the National Academy of Sciences.

[46]  Ralph Weissleder,et al.  Both p16(Ink4a) and the p19(Arf)-p53 pathway constrain progression of pancreatic adenocarcinoma in the mouse. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[47]  A. Rust,et al.  Sleeping Beauty mutagenesis reveals cooperating mutations and pathways in pancreatic adenocarcinoma , 2012, Proceedings of the National Academy of Sciences.

[48]  D. Tuveson,et al.  Gamma secretase inhibition promotes hypoxic necrosis in mouse pancreatic ductal adenocarcinoma , 2012, The Journal of experimental medicine.

[49]  G. Elder,et al.  Animal transgenesis: an overview , 2010, Brain Structure and Function.

[50]  M. Barbacid,et al.  Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. , 2007, Cancer cell.

[51]  Dissecting the Cellular Origins of Pancreatic Cancer , 2006, Cell cycle.

[52]  T. Jacks,et al.  Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras. , 2001, Genes & development.

[53]  H. Ku Minireview: pancreatic progenitor cells--recent studies. , 2008, Endocrinology.

[54]  J. Cameron,et al.  Multicomponent Analysis of the Pancreatic Adenocarcinoma Progression Model Using a Pancreatic Intraepithelial Neoplasia Tissue Microarray , 2003, Modern Pathology.

[55]  Gerald C. Chu,et al.  Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. , 2009, Cancer cell.

[56]  Carlos Cuevas,et al.  Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. , 2012, Cancer cell.

[57]  D. Tuveson,et al.  K-Ras-driven pancreatic cancer mouse model for anticancer inhibitor analyses. , 2008, Methods in enzymology.

[58]  C. Der,et al.  Inhibition of Ras for cancer treatment: the search continues. , 2011, Future medicinal chemistry.

[59]  G. Parmigiani,et al.  Core Signaling Pathways in Human Pancreatic Cancers Revealed by Global Genomic Analyses , 2008, Science.

[60]  M. Korc,et al.  Deletion of Rb accelerates pancreatic carcinogenesis by oncogenic Kras and impairs senescence in premalignant lesions. , 2011, Gastroenterology.

[61]  David Allard,et al.  Inhibition of Hedgehog Signaling Enhances Delivery of Chemotherapy in a Mouse Model of Pancreatic Cancer , 2009, Science.

[62]  M. Korc,et al.  The Nestin progenitor lineage is the compartment of origin for pancreatic intraepithelial neoplasia , 2007, Proceedings of the National Academy of Sciences.

[63]  Gerald C. Chu,et al.  Oncogenic Kras Maintains Pancreatic Tumors through Regulation of Anabolic Glucose Metabolism , 2012, Cell.

[64]  D. Tuveson,et al.  nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. , 2012, Cancer discovery.

[65]  M. Barbacid,et al.  What we have learned about pancreatic cancer from mouse models. , 2012, Gastroenterology.

[66]  J. Deng,et al.  Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine , 2011, Nature Genetics.

[67]  D. Tuveson,et al.  Dynamics of the immune reaction to pancreatic cancer from inception to invasion. , 2007, Cancer research.

[68]  J. Feramisco,et al.  The product of ras is a GTPase and the T24 oncogenic mutant is deficient in this activity , 1984, Nature.

[69]  Paul Timpson,et al.  Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer , 2010, Proceedings of the National Academy of Sciences.

[70]  M. Hollingsworth,et al.  Sonic hedgehog paracrine signaling regulates metastasis and lymphangiogenesis in pancreatic cancer , 2009, Oncogene.

[71]  R. Hruban,et al.  Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. , 2003, Cancer cell.

[72]  D. Bar-Sagi,et al.  Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. , 2012, Cancer cell.

[73]  Gerald C. Chu,et al.  GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. , 2009, Genes & development.

[74]  F. C. Pan,et al.  Pancreas organogenesis: From bud to plexus to gland , 2011, Developmental dynamics : an official publication of the American Association of Anatomists.

[75]  David J. Anderson,et al.  Notch signalling controls pancreatic cell differentiation , 1999, Nature.

[76]  Jason R. Gunn,et al.  Acute Pancreatitis Accelerates Initiation and Progression to Pancreatic Cancer in Mice Expressing Oncogenic Kras in the Nestin Cell Lineage , 2011, PloS one.

[77]  T. Schoeb,et al.  Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. , 2007, Cancer research.

[78]  R. Hruban,et al.  Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. , 2007, Cancer cell.

[79]  Gerald C. Chu,et al.  PTEN is a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates an NF-κB-cytokine network. , 2011, Cancer discovery.

[80]  T. Ludwig,et al.  BRCA1 Tumor Suppression Depends on BRCT Phosphoprotein Binding, But Not Its E3 Ligase Activity , 2011, Science.

[81]  Todd E. Scheetz,et al.  Somatic Mutagenesis with a Sleeping Beauty Transposon System Leads to Solid Tumor Formation in Zebrafish , 2011, PloS one.

[82]  Ashok R Venkitaraman,et al.  Germline Brca2 heterozygosity promotes Kras(G12D) -driven carcinogenesis in a murine model of familial pancreatic cancer. , 2010, Cancer cell.

[83]  J. Norman,et al.  Mutant p53 Drives Invasion by Promoting Integrin Recycling , 2009, Cell.

[84]  G. Kristiansen,et al.  The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma , 2012, Nature.

[85]  Gerald C. Chu,et al.  Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. , 2006, Genes & development.

[86]  Mallika Singh,et al.  Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models , 2010, Nature Biotechnology.

[87]  L. C. Murtaugh,et al.  Pancreatic stem cells , 2008 .

[88]  Malte Buchholz,et al.  Stromal biology and therapy in pancreatic cancer , 2010, Gut.

[89]  R. Rad,et al.  Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma , 2010, Proceedings of the National Academy of Sciences.

[90]  D. Tuveson,et al.  Suppression of BRAF(V599E) in human melanoma abrogates transformation. , 2003, Cancer research.

[91]  Brian Bierie,et al.  Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma. , 2011, The Journal of clinical investigation.

[92]  J. Kench,et al.  Stabilization of beta-catenin induces pancreas tumor formation. , 2008, Gastroenterology.

[93]  R. Hruban,et al.  Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. , 2005, Cancer cell.

[94]  A. Maitra,et al.  Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.