Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Using mice to unveil the genetics of cancer resistance

2012
...Read more
Review Using mice to unveil the genetics of cancer resistance Louise van der Weyden , David J. Adams Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton,Cambridge, CB10 1SA, UK a b s t r a c t a r t i c l e i n f o Article history: Received 6 March 2012 Received in revised form 10 May 2012 Accepted 13 May 2012 Available online 19 May 2012 Keywords: Cancer Resistance Tumorigenesis Mouse In the UK, four in ten people will develop some form of cancer during their lifetime, with an individual's rel- ative risk depending on many factors, including age, lifestyle and genetic make-up. Much research has gone into identifying the genes that are mutated in tumorigenesis with the over-whelming majority of genetically- modified (GM) mice in cancer research showing accelerated tumorigenesis or recapitulating key aspects of the tumorigenic process. Yet if six out of ten people will not develop some form of cancer during their life- time, together with the fact that some cancer patients experience spontaneous regression/remission, it sug- gests there are ways of ‘resisting’ cancer. Indeed, there are wildtype, spontaneously-arising mutants and GM mice that show some form of ‘resistance’ to cancer. Identification of mice with increased resistance to cancer is a novel aspect of cancer research that is important in terms of providing both chemopreventative and ther- apeutic options. In this review we describe the different mouse lines that display a ‘cancer resistance’ pheno- type and discuss the molecular basis of their resistance. © 2012 Elsevier B.V. All rights reserved. Contents 1. Introduction .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313 2. Cancer resistance in wildtype mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313 2.1. Wildtype mouse strains show differential susceptibility/resistance to cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313 2.2. Exploiting genetic diversity between wildtype mouse strains to identify ‘resistance’ genes . . . . . . . . . . . . . . . . . . . . . . 314 2.3. Heterocephalus glaber . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314 3. Cancer resistance in spontaneously-arising mutant mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314 3.1. Dm1 mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314 3.2. S-27,S-31 and S-87/2 mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314 3.3. SR/CR mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315 4. Cancer resistance in genetically-modified (GM) mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315 4.1. T-cell signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315 4.1.1. Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 315 4.1.2. Tumor necrosis factors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316 4.1.3. CTLA-4 .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 316 4.2. Inflammation .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317 4.2.1. Phospholipase A 2 enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317 4.2.2. Cyclooxygenase enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317 Biochimica et Biophysica Acta 1826 (2012) 312–330 Abbreviations: AC, adenocarcinoma; AOM, azoxymethane; APC, antigen-presenting cells; BH, BCL2 homology; CCl 4 , carbon tetrachloride; cM, centimorgan; COX, cyclo-oxygenase; CTLA-4, cytotoxic T-lymphocyte antigen 4; DC, dendritic cells; DEN, diethylnitrosamine; DMBA, 7,12-dimethylbenz[α]anthracene; DSB, DNA-strand break; DSS, dextran sodium G5, five generations of inter-crossing (late generations); GM, genetically modified; IFN-γ, interferon-gamma; IκB, IkappaB; IKK, IκB kinase; IL, interleukin; Mb, megabase; MCP-1 cyte chemoattractantprotein-1; MEF, mouse embryonicfibroblasts;MHC, major histocompatibility;MMTV, mouse mammary tumor virus; MNNG, N-methyl-N′-nitro-N- nitrosoguanidine; MNU, N-Nitroso-N-methylurea; Mom, modifier of Min; NER, nucleotide excision repair; NF-κB, nuclear factor kappa B; NMR, naked mole-rat; NO, nitric oxide; N nitric oxide synthase; PA, pulmonary adenomas; Par1, pulmonary adenoma resistance 1; PI3K, phosphoinositide 3-kinase; PI3P, phosphatidylinositol 3-phosphate; PLA 2 , phospholipase A2; PUFA, polyunsaturated fatty acids; RASSF, Ras association family; RBD, Ras binding domain; R-Smads, receptor-activated Smads; SAC, selective for apoptosis of cancer cell cell carcinoma; SCID, severe combined immunodeficiency; SQ, squamous cell carcinoma; SR/CR, spontaneous regression/complete resistance; TAg, T antigen; TERT, telomeras transcriptase; TGF, transforming growth factor; Th cell, T helper cell; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; TPA, 12-O-tetradecanoylphorbol-13-acet telomerase RNA; UC, ulcerative colitis; UTR, untranslated region; UVB, ultraviolet B ⁎ Corresponding author. Tel.: + 44 1223 834 244; fax: +44 1223 496 802. E-mail address: lvdw@sanger.ac.uk (L. van der Weyden). 0304-419X/$ – see front matter © 2012 Elsevier B.V. All rights reserved. doi:10.1016/j.bbcan.2012.05.003 Contents lists available at SciVerse ScienceDirect Biochimica et Biophysica Acta j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / b b a c a n
4.2.3. Nitric oxide synthases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319 4.2.4. IkappaB kinase complex. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319 4.2.5. Polyunsaturated fatty acids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319 4.3. Cell growth/death . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320 4.3.1. p53 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320 4.3.2. INK4a/ARF/INK4b locus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320 4.3.3. Phosphatase and tensin homologue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321 4.3.4. Activator protein 1 .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321 4.3.5. Transforming growth factor-β . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321 4.3.6. Smad3 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321 4.3.7. Bone morphogenetic proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322 4.3.8. B-cell lymphoma 2 family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322 4.3.9. Protein tyrosine phosphatase 1B . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322 4.3.10. Rassf3 .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322 4.3.11. Serine/threonine protein kinases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.3.12. GATA factors .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.3.13. Prostate apoptosis response-4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.4. Maintaining genomic stability/integrity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.4.1. PPM1D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.4.2. Poly(ADP-ribose) polymerase-1 .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 323 4.4.3. O 6 -methylguanine-DNA methyltransferase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324 4.4.4. DNA methyltransferase 1 .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324 4.4.5. Terc . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 324 4.5. Miscellaneous . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325 4.5.1. Eph receptor A2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325 4.5.2. Fibroblast factor binding protein. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325 5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325 Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326 Acknowledgements .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326 References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326 1. Introduction There is no doubt that cancer is a devastating disease, with four out of every ten people being struck by cancer atsome point in their life.However,the flip-side of this fact is less emphasized, spe- cifically that six in every ten people will not develop cancer. When taken together with the fact that many heavy smokers remain can- cer free (~ 10% smokers develop lung cancer, depending upon how much they have smoked) and some cancer patients experience spontaneous regression/remission of their tumor [1], this suggests that ‘cancer resistance’ is possible.The germline transmission ofa defective allele at the tumor suppressor locus greatly increases the risk of developing certain forms of cancer, however,the major- ity of cancers are sporadic,and epidemiologicaldata suggestthat there are likely to be multiple low-penetrance genes that segregate in the human population and confer strong resistance or suscepti- bility to environmentally-induced cancers [2]. It has been proposed that individual cancer risk is determined by combinations of resis- tance or susceptibility alleles inherited through the germline, and while most humans will be at average risk, some might inherit more than their fair share of susceptibility alleles,and be highly cancer prone, while others will inherit a predominance of resis- tance alleles and be very resistant [3]. Animal models of cancer are commonly used to study tumor biol- ogy and develop new approaches to conquering human cancer. Mice are widely used as the preeminent animal models in cancer research, due to their small size, inbred nature,and underlying similarities in the biology of tumorigenesis with humans, implying that genes that control susceptibility/resistance to tumor development in mice will also be relevant to the human situation. Thus the development and characterization ofmice models displaying a cancer resistant phenotype are crucial for the identification of genes involved in cancer resistance and offer invaluable tools for determining potential chemopreventive strategies. 2. Cancer resistance in wildtype mice 2.1. Wildtype mouse strains show differential susceptibility/resistance to cancer An important feature of mouse tumorigenesis is strain-dependent tumor susceptibility/resistance. Wildtype mouse strains show differ- ential susceptibility/resistance to a variety of both spontaneous and chemically-induced tumor types. For example,compared to the es- sentially lung tumor resistant C57BL/6J strain, A/J mice exhibit high susceptibility to spontaneous and chemically-induced lung tumors [4].Similarly,C57BL/6J mice are also resistant to chemically-induced liver tumors [5]. For the organotropic colon carcinogen azoxymethane (AOM), A/J and SWR/J strains are susceptible,whereas the AKR/J strain is resistant [6]. For viral-induced oncogenesis in mice, several host genes are known to play a major role in determining the level of resistance/susceptibility including the complex genetic locus cod- ing for the major histocompatibility (MHC) antigens ofthe mouse (the H-2 locus) (reviewed in Ref. [7]). For example,in infections with Gross leukemia virus [8,9], Tennant leukemia virus (B/T-L) [10], some strains of mammary tumor virus [11],and Friend leukemia virus [12], H-2 b/b is strongly associated with resistance to oncogenesis, whereas H-2 a/a , H-2 d/d , and H-2 k/k are associated with susceptibility to oncogenesis. The X/Gf strain has a very low incidence of spontaneous and in- duced (chemical, radiation) tumors, believed to be due to their highly endogenous immune competence, and an absence (or very low pres- ence) of any leukemia or mammary tumor viruses [13]. Similarly, wild-derived mouse strains (such as Musculus spretus and Musculus castaneous), derived primarily from captured wild mice that have been bred to homozygosity through sibling matings, and are thus from populations with much greater diversity than found in the clas- sical laboratory strains [14], are often more resistant to carcinogens and pathogens than the commonly used laboratory-derived strains. 313 L.van der Weyden, D.J.Adams / Biochimica et Biophysica Acta 1826 (2012) 312330
Biochimica et Biophysica Acta 1826 (2012) 312–330 Contents lists available at SciVerse ScienceDirect Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbacan Review Using mice to unveil the genetics of cancer resistance Louise van der Weyden ⁎, David J. Adams Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK a r t i c l e i n f o Article history: Received 6 March 2012 Received in revised form 10 May 2012 Accepted 13 May 2012 Available online 19 May 2012 Keywords: Cancer Resistance Tumorigenesis Mouse a b s t r a c t In the UK, four in ten people will develop some form of cancer during their lifetime, with an individual's relative risk depending on many factors, including age, lifestyle and genetic make-up. Much research has gone into identifying the genes that are mutated in tumorigenesis with the over-whelming majority of geneticallymodified (GM) mice in cancer research showing accelerated tumorigenesis or recapitulating key aspects of the tumorigenic process. Yet if six out of ten people will not develop some form of cancer during their lifetime, together with the fact that some cancer patients experience spontaneous regression/remission, it suggests there are ways of ‘resisting’ cancer. Indeed, there are wildtype, spontaneously-arising mutants and GM mice that show some form of ‘resistance’ to cancer. Identification of mice with increased resistance to cancer is a novel aspect of cancer research that is important in terms of providing both chemopreventative and therapeutic options. In this review we describe the different mouse lines that display a ‘cancer resistance’ phenotype and discuss the molecular basis of their resistance. © 2012 Elsevier B.V. All rights reserved. Contents 1. 2. 3. 4. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cancer resistance in wildtype mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1. Wildtype mouse strains show differential susceptibility/resistance to cancer . . . . . . 2.2. Exploiting genetic diversity between wildtype mouse strains to identify ‘resistance’ genes 2.3. Heterocephalus glaber . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cancer resistance in spontaneously-arising mutant mice . . . . . . . . . . . . . . . . . . . 3.1. Dm1 mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2. S-27, S-31 and S-87/2 mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3. SR/CR mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Cancer resistance in genetically-modified (GM) mice . . . . . . . . . . . . . . . . . . . . . 4.1. T-cell signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1.1. Cytokines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1.2. Tumor necrosis factors . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.1.3. CTLA-4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2. Inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4.2.1. Phospholipase A2 enzymes . . . . . . . . . . . . . . . . . . . . . . . . . 4.2.2. Cyclooxygenase enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313 313 313 314 314 314 314 314 315 315 315 315 316 316 317 317 317 Abbreviations: AC, adenocarcinoma; AOM, azoxymethane; APC, antigen-presenting cells; BH, BCL2 homology; CCl4, carbon tetrachloride; cM, centimorgan; COX, cyclo-oxygenase; CTLA-4, cytotoxic T-lymphocyte antigen 4; DC, dendritic cells; DEN, diethylnitrosamine; DMBA, 7,12-dimethylbenz[α]anthracene; DSB, DNA-strand break; DSS, dextran sodium sulfate; G5, five generations of inter-crossing (late generations); GM, genetically modified; IFN-γ, interferon-gamma; IκB, IkappaB; IKK, IκB kinase; IL, interleukin; Mb, megabase; MCP-1, monocyte chemoattractant protein-1; MEF, mouse embryonic fibroblasts; MHC, major histocompatibility; MMTV, mouse mammary tumor virus; MNNG, N-methyl-N′-nitro-Nnitrosoguanidine; MNU, N-Nitroso-N-methylurea; Mom, modifier of Min; NER, nucleotide excision repair; NF-κB, nuclear factor kappa B; NMR, naked mole-rat; NO, nitric oxide; NOS, nitric oxide synthase; PA, pulmonary adenomas; Par1, pulmonary adenoma resistance 1; PI3K, phosphoinositide 3-kinase; PI3P, phosphatidylinositol 3-phosphate; PLA2, phospholipase A2; PUFA, polyunsaturated fatty acids; RASSF, Ras association family; RBD, Ras binding domain; R-Smads, receptor-activated Smads; SAC, selective for apoptosis of cancer cells; SCC, small cell carcinoma; SCID, severe combined immunodeficiency; SQ, squamous cell carcinoma; SR/CR, spontaneous regression/complete resistance; TAg, T antigen; TERT, telomerase reverse transcriptase; TGF, transforming growth factor; Th cell, T helper cell; TNF, tumor necrosis factor; TNFR, tumor necrosis factor receptor; TPA, 12-O-tetradecanoylphorbol-13-acetate; TR, telomerase RNA; UC, ulcerative colitis; UTR, untranslated region; UVB, ultraviolet B ⁎ Corresponding author. Tel.: + 44 1223 834 244; fax: +44 1223 496 802. E-mail address: lvdw@sanger.ac.uk (L. van der Weyden). 0304-419X/$ – see front matter © 2012 Elsevier B.V. All rights reserved. doi:10.1016/j.bbcan.2012.05.003 313 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 4.2.3. Nitric oxide synthases . . . . . . . . . 4.2.4. IkappaB kinase complex . . . . . . . . 4.2.5. Polyunsaturated fatty acids . . . . . . . 4.3. Cell growth/death . . . . . . . . . . . . . . . 4.3.1. p53 . . . . . . . . . . . . . . . . . . 4.3.2. INK4a/ARF/INK4b locus . . . . . . . . . 4.3.3. Phosphatase and tensin homologue . . . 4.3.4. Activator protein 1 . . . . . . . . . . . 4.3.5. Transforming growth factor-β . . . . . . 4.3.6. Smad3 . . . . . . . . . . . . . . . . 4.3.7. Bone morphogenetic proteins . . . . . . 4.3.8. B-cell lymphoma 2 family . . . . . . . 4.3.9. Protein tyrosine phosphatase 1B . . . . 4.3.10. Rassf3 . . . . . . . . . . . . . . . . . 4.3.11. Serine/threonine protein kinases . . . . 4.3.12. GATA factors . . . . . . . . . . . . . . 4.3.13. Prostate apoptosis response-4 . . . . . . 4.4. Maintaining genomic stability/integrity . . . . . 4.4.1. PPM1D . . . . . . . . . . . . . . . . 4.4.2. Poly(ADP-ribose) polymerase-1 . . . . . 4.4.3. O 6-methylguanine-DNA methyltransferase 4.4.4. DNA methyltransferase 1 . . . . . . . . 4.4.5. Terc . . . . . . . . . . . . . . . . . . 4.5. Miscellaneous . . . . . . . . . . . . . . . . . 4.5.1. Eph receptor A2 . . . . . . . . . . . . 4.5.2. Fibroblast factor binding protein . . . . 5. Conclusions . . . . . . . . . . . . . . . . . . . . . . Conflict of interest . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319 319 319 320 320 320 321 321 321 321 322 322 322 322 323 323 323 323 323 323 324 324 324 325 325 325 325 326 326 326 1. Introduction 2. Cancer resistance in wildtype mice There is no doubt that cancer is a devastating disease, with four out of every ten people being struck by cancer at some point in their life. However, the flip-side of this fact is less emphasized, specifically that six in every ten people will not develop cancer. When taken together with the fact that many heavy smokers remain cancer free (~ 10% smokers develop lung cancer, depending upon how much they have smoked) and some cancer patients experience spontaneous regression/remission of their tumor [1], this suggests that ‘cancer resistance’ is possible. The germline transmission of a defective allele at the tumor suppressor locus greatly increases the risk of developing certain forms of cancer, however, the majority of cancers are sporadic, and epidemiological data suggest that there are likely to be multiple low-penetrance genes that segregate in the human population and confer strong resistance or susceptibility to environmentally-induced cancers [2]. It has been proposed that individual cancer risk is determined by combinations of resistance or susceptibility alleles inherited through the germline, and while most humans will be at average risk, some might inherit more than their fair share of susceptibility alleles, and be highly cancer prone, while others will inherit a predominance of resistance alleles and be very resistant [3]. Animal models of cancer are commonly used to study tumor biology and develop new approaches to conquering human cancer. Mice are widely used as the preeminent animal models in cancer research, due to their small size, inbred nature, and underlying similarities in the biology of tumorigenesis with humans, implying that genes that control susceptibility/resistance to tumor development in mice will also be relevant to the human situation. Thus the development and characterization of mice models displaying a cancer resistant phenotype are crucial for the identification of genes involved in cancer resistance and offer invaluable tools for determining potential chemopreventive strategies. 2.1. Wildtype mouse strains show differential susceptibility/resistance to cancer An important feature of mouse tumorigenesis is strain-dependent tumor susceptibility/resistance. Wildtype mouse strains show differential susceptibility/resistance to a variety of both spontaneous and chemically-induced tumor types. For example, compared to the essentially lung tumor resistant C57BL/6J strain, A/J mice exhibit high susceptibility to spontaneous and chemically-induced lung tumors [4]. Similarly, C57BL/6J mice are also resistant to chemically-induced liver tumors [5]. For the organotropic colon carcinogen azoxymethane (AOM), A/J and SWR/J strains are susceptible, whereas the AKR/J strain is resistant [6]. For viral-induced oncogenesis in mice, several host genes are known to play a major role in determining the level of resistance/susceptibility including the complex genetic locus coding for the major histocompatibility (MHC) antigens of the mouse (the H-2 locus) (reviewed in Ref. [7]). For example, in infections with Gross leukemia virus [8,9], Tennant leukemia virus (B/T-L) [10], some strains of mammary tumor virus [11], and Friend leukemia virus [12], H-2 b/b is strongly associated with resistance to oncogenesis, whereas H-2 a/a, H-2 d/d, and H-2 k/k are associated with susceptibility to oncogenesis. The X/Gf strain has a very low incidence of spontaneous and induced (chemical, radiation) tumors, believed to be due to their highly endogenous immune competence, and an absence (or very low presence) of any leukemia or mammary tumor viruses [13]. Similarly, wild-derived mouse strains (such as Musculus spretus and Musculus castaneous), derived primarily from captured wild mice that have been bred to homozygosity through sibling matings, and are thus from populations with much greater diversity than found in the classical laboratory strains [14], are often more resistant to carcinogens and pathogens than the commonly used laboratory-derived strains. 314 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 Table 1 Major tumor resistance loci identified from crosses between ‘susceptible’ and ‘resistant’ mouse strains. AC, adenocarcinomas; PA, pulmonary adenomas; Par-1, pulmonary adenoma resistance-1; SCC, small cell carcinoma; SQ, squamous cell carcinoma. Resistance locus F1 mice (back-crossed to perform the linkage analysis) Details of the locus Chromosomes 5 and 7: skin cancer NIH × M. spretus (NIH mice are very sensitive to DMBA/TPA-induced skin papillomas and carcinomas, whereas M. spretus are resistant) A/J × M. spretus (A/J mice are very sensitive to urethane-induced PA, whereas M. spretus are resistant) A/J × SM/J (SM/J mice are resistant to urethane-induced PA) A single locus on chromosome 5 affecting both early and late stages of malignancy, and two independent loci on chromosome 7 primarily affecting benign tumor development (not progression) [236]. Chromosome 11: lung cancer Chromosome 18: lung cancer Chromosome 12: lung cancer A/J × BALB/cByJ (BALB/cByJ mice are 14 times more resistant to urethane-induction of PA than A/J mice) A/J × SM/J 2.2. Exploiting genetic diversity between wildtype mouse strains to identify ‘resistance’ genes An approach to identify these resistance loci has been to exploit the genetic diversity between the ‘resistant’ and ‘susceptible’ mouse strains. For example, crosses between M. spretus and inbred laboratory strains showed that M. spretus/M. musculus F1 hybrids were extremely resistant to most chemical carcinogenesis protocols, failing to develop appreciable numbers of tumors of a wide range of tissues [15]. Such interspecific crosses have led to the detection of multiple loci affecting tumor development, such as the mapping of several loci that control resistance to different stages of skin tumor development and resistance genes for lung tumor development (see Table 1). Although undeniably such breeding strategies have been predominantly used to identify tumor ‘susceptibility’ loci (summarized in Ref. [3]), it is important to remember that genes classified as conferring ‘resistance’ in a particular mouse strain can also reveal themselves as susceptibility loci in subsequent crosses into other genetic backgrounds (and presumably vise versa) [16]. Thus individual loci might only be revealed as resistance or susceptibility loci after crossing to a genetic background that allows gene–gene interactions resulting in the expression of the trait, as inter-locus interactions have been shown to be an important component of tumor susceptibility [17,18]. 2.3. Heterocephalus glaber A rodent that has been shown to possess natural resistance to cancer is the naked mole-rat (NMR; Heterocephalus glaber). In a colony of 1500 animals in captivity, the oldest individuals are more than 30 years of age, and although animals over 24 years show evidence of frailty and die of age-related causes, death because of cancer has never been observed, and necropsies have not revealed incidental tumors [19]. Thus, the NMR appears to be a cancer-resistant mammal. In vitro studies on NMR fibroblast cells found that they react to signals in the culture environment that cause them to express unusually high levels of the tumor suppressor p16 INK4A, and expression of the combination of the oncogenes SV40 large T antigen (TAg) and Ras G12V reduced the level of p16 INK4A, thereby permitting the cells to grow well in culture but did not confer the ability to grow as colonies in soft agarose (a surrogate assay for tumorigenicity) [20]. More recently, NMR cells expressing SV40 TAg and Ras oncogenes were shown to rapidly enter crisis when transplanted into SCID mice and thus unable to form tumors [21]. Thus understanding the basis for Par-1 maps to a 2 cM (2.2 Mb) region between D11Mit70 and Hoxb9. Candidate gene is Tob1 [237,238]. Par-1 corresponds to human 17q11–23. LOH at 17q was found in 53% of AC (but only at low levels in the other lung cancer subtypes, suggesting that the gene is selective for AC, probably at the level of the target cell) [239,240]. Par-2 maps to a 0.5 cM region between D18Mit103 and D18Mit188 [241–243]. Par-3 candidate gene is protein kinase C eta (nPKCη), which is expressed exclusively in skin and lung and down-regulated in PA. Par1 and Par3 act synergistically. Par-3 corresponds to human 14q11–24. LOH at 14q was found in 30–42% of AC, SQ and SCC (affecting all 3 subtypes of lung cancer suggests that it is related to the progression of lung tumors in general) [239,240]. cancer resistance in the NMR will provide important clues for mechanisms defining the causes of cancer susceptibility and resistance in mammals. 3. Cancer resistance in spontaneously-arising mutant mice Most inbred strains of mice succumb to growing malignant tumors, including both endogenously-arising ones (autochthonous tumors) and transplantable tumors (non-autochthonous tumors) that originated in a mouse of the same strain. However, as with any biological function, this phenotype can change through a mutation, and although spontaneous gene mutations for resistance (or increased susceptibility) to a malignant tumor are rare events, they provide valuable information about the biological function of the gene involved. Mouse mutations for resistance to transplantation of a malignant tumor or its metastases are discussed in more detail below. 3.1. Dm1 mice Studies to identify spontaneous mutants of the mouse MHC locus (H-2 mutations) were initiated in the early 1960s using skin graft survival or rejection as a screening tool for mutants [22,23]. For example, H-2 dm1 is a spontaneous MHC class I mutation first identified in a mouse of the B10.D2 strain (MHC haplotype H-2 d) because it rejected parental B10.D2 skin grafts [22]. Later studies showed that this mutation was due to a hybrid gene containing the 5′ part of the D d gene and the 3′ part of the L d gene, resulting in a deletion of the segment between these genes that contains three other MHC class I genes [24]. Subsequently, the heterozygous carriers of the dm1 mutation (H-2 dm1/H-2 b) were found to recover from Friend retrovirusinduced leukemia, whereas normal heterozygous mice (H-2 d/H-2 b) die from the disease [25]. Unfortunately, the immunological mechanism of spontaneous regression of these tumors in H-2 dm1/H-2 b mice has not been characterized in full detail. 3.2. S-27, S-31 and S-87/2 mice In screens designed to expose host genes for resistance, mice were analysed for their ability to survive transplantable metastatic tumors [26,27]. In a screen of over 70,000 mice, three spontaneous mutations affecting the incidence of survival and metastases were identified. One survivor of the C57BL/10Sn strain, designated S-27, survived an intraperitoneal injection of EL4 lymphoma cells (H-2 b), which normally kills C57BL/10Sn mice (H-2 b). When the mutant gene was L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 crossed onto the C57BL/6J background (H-2 b), the S-27 mutants were able to resist moderate (but not high) doses of transplantable B6 tumor, and resist metastasis to a variety of cell lines (including radiation-induced lymphoma BB, chemically-induced lymphoma EL4, and rhabdomyosarcoma MCA/77-23, which typically result in metastasis and death in wildtype C57BL/6J mice), 1 indicating that S27 mice carry a gene for resistance to metastasis [27,28]. In contrast, the S-31 mutant identified in the same screen resists the initial EL4 innoculum but is highly susceptible to its metastasis. Similarly, the S-87/2 mutant was identified in a screen using the C57BL/6J strain for susceptibility to sarcoma MCA/77-23 cells, and although it is resistant to the original tumor transplant, it shows increased susceptibility to metastasis. Genes for resistance or susceptibility to the tumor and its metastasis in these three S-mutants were found to be in novel single exon genes Aβ4–7 comprising the Aβ6 gene family, whose protein coding region is very similar to the classical H-2-Ab cDNA encoding the β chain of MHC class II molecules, found on the surface of antigen presenting cells (APC). However, these mutant Aβ4–6 genes have been shown to be molecularly unstable in somatic cells of mice carrying them, including the mutant strains S-27 (genes Aβ4 b, Aβ6 w302), S31 (hybrid between Aβ5 s5 and Aβ4 b genes) and S-87/2 (gene not yet identified), and this instability results in somatic mosaicism of these nontrivial MHC class II molecules in their APC [29]. Thus these genes controlling susceptibility/resistance to the spread of metastatic tumors are remarkably different from the classical MHC gene system. 315 sentinels of the immune response and T cells as effectors of adaptive anti-tumor immunity, with a variety of cytotoxic molecules mediating the critical role of host prevention of malignancy. Conversely, while acute inflammation is a part of the defense response, chronic inflammation can lead to a wide variety of diseases, including cancer, and in the last decade a growing body of epidemiological and clinical data has emerged to support the concept that longstanding inflammation potentiates or promotes tumor development, growth and progression. The balance between cell proliferation and apoptosis is influenced by genes that contribute to the development of cancer (oncogenes) and those that encode proteins that normally suppress tumor formation (tumor suppressor genes), and virtually all cancer cells contain mutations that enable evasion of apoptosis through dynamic interplays between activated oncogenes and/or mutated tumor suppressor genes. Genetic stability tumor suppressor genes preserve genome integrity, and activate the numerous multiple-step DNA repair pathways that exist, depending on the nature of the DNA lesion. Thus genes that augment the host immune system; and/or dampen down the inflammatory response; and/or halt proliferation and promote apoptosis of the tumor cells; and/or maintain the integrity of the genome all play important roles in cancer resistance. Examples of such genes and their modifications in mouse models of cancer are discussed in more detail below and summarized in Table 2. Unfortunately, some of these GM ‘cancer resistant’ mice also show associated phenotypes such as reduced lifespan, susceptibility to pathogens and nephropathy (reviewed in Ref. [35]). 3.3. SR/CR mice 4.1. T-cell signaling In 2003, Cui and colleagues discovered a single mouse in their BALB/c colony that displayed resistance to intraperitoneal injection of murine sarcoma S180 cells (H-2 q), 2 which typically results in ascites accumulation and death within 3–4 weeks [30]. When this mouse was bred with wildtype S180-sensitive mice, of a variety of different genetic backgrounds, 30–40% of the F1 progeny was also resistant, and this dominant trait was termed ‘spontaneous regression/ complete resistance’ (SR/CR) [30,31]. Interestingly, the response does vary according to age, with SR/CR mice that are 6 weeks old on first exposure to S180 cells showing complete resistance to ascites formation, whereas mice that are 22 weeks old on first exposure showing development of ascites accumulation over the first 2 weeks, which then disappears within 24 h [30,31]. In addition to resistance to S180 cells, SR/CR mice either on a BALB/c or C57BL/6J congenic background show significantly higher levels of resistance to a wide variety of cancer cell lines in comparison to wildtype mice (including MethA sarcoma, B16 melanoma, LL/2 lung carcinoma and J774 and EL-4 lymphoma cells) [32]. The SR/CR trait of cancer resistance is mediated primarily by leukocytes of the innate immune system [33,34] and is based on two separate processes: leukocyte migration/infiltration to the site of the cancer cells and recognition of common surface properties on cancer cells (Fig. 1) [32]. The identity of the chromosomal region responsible for this genetic trait is as yet unknown. 4. Cancer resistance in genetically-modified (GM) mice The idea that the immune system might help protect humans against cancer was first suggested by Paul Ehrlich over a century ago, and much research has focused upon dendritic cells (DCs) as 1 Spontaneous metastasis assay is when intradermally growing tumors are surgically removed and mice scored for metastases that occur afterwards. In susceptible C57BL/6J mice, surgery does not prevent the metastases, which grow in various organs and inevitably kill the mouse. 2 Mouse S180 sarcoma cells do not show detectable levels of MHC protein on their cell surface and thus can form highly aggressive cancers in all strains of laboratory mouse and rats. 4.1.1. Cytokines Cytokines are hormonal messengers responsible for most of the biological effects in the immune system, and can be functionally divided into two groups: pro-inflammatory and anti-inflammatory (but promote allergic responses). T lymphocytes are a major source of cytokines, the most prolific of which are those expressing CD4 (also known as helper T cells; Th), and this subset can be further subdivided into Th1 and Th2. Th1-type cytokines (particularly IFN-γ) tend to produce pro-inflammatory responses, whereas Th2-type cytokines (such as IL-4, 5, 10 and 13) tend to produce more of an anti-inflammatory response. Despite the fact that excessive proinflammatory responses can lead to uncontrolled tissue damage, cytokines released from Th1 cells are in particularly effective in shifting immune systems into a Th1-dominant state, which enhances cellmediated immunity [36]. The Th1 branch of the immune system, which employs T cells, NK cells and macrophages, plays major role in combating cancer but growing cancers actively suppress immune responses and disregulate the activity of these effector cells [37]. Chemokines are a family of small cytokines that have the ability to induce directed chemotaxis in nearby responsive cells (‘chemotactic cytokines’). Members of the chemokine family are divided into four groups depending on the spacing of their first two cysteines: CC (or β-chemokine), CXC (or α-chemokines), C (or γ chemokines), and CX3C (or δ-chemokines). Some chemokines are pro-inflammatory and can be induced during an immune response to recruit cells of the immune system to a site of infection, while others are homeostatic and involved in controlling the migration of cells during normal processes of tissue maintenance or development. A number of studies transplant/xenograft models in mice have demonstrated that secretion of cytokines and chemokines from tumors can produce anti-tumor effects (some examples of which are shown in Table 3). Although further investigation is required to elucidate the precise mechanisms involved in the anti-tumor activity of these Th1 cytokines/chemokines, studies using mouse models have helped determine that in some cases it can be NK-dependent. For example, depletion of NK cells from nude mice with anti-asialo GM1 316 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 A B C Fig. 1. SR/CR leukocyte migration and destruction of cancer cells. Leukocyte infiltration is an important part of the SR/CR resistance mechanism [32]. (A) S180 sarcoma cells (large orange circles) secrete chemotactic factors (small orange circles) that attract SR/CR leukocytes (large blue circles), allowing tight contact between the leukocytes and cancer cells, which facilitates their destruction. (B) In contrast, other cancer cell lines, such as LL/2 and MethA (large yellow circles) produce little or no chemoattractant and are therefore not efficiently killed by SR/CR leukocytes. (C) However, co-injection of MethA or LL/2 cells with S180 cells results in attraction of SR/CR leukocytes and efficient destruction of these cancer cells. antibody diminishes the growth retardation of IL-27-transfected colon cancer (Colon 26/IL-27) tumors and IL-21-transfected pancreatic cancer (AsPC-1/IL-21) tumors [38]. Furthermore, the use of immunodeficient mouse lines (such as nude and SCID mice) has helped to determine that there is also involvement of specific T-cell subsets in this process. For example, while the growth of AsPC-1/IL21 and AsPC-1/IL-23 tumors in nude mice was retarded compared with that of parental (untransfected AsPC-1) tumors, the growth of AsPC-1/IL-23 tumors in SCID mice was not different from that of parental tumors [39]. In addition to the transplant/xenograft models as mentioned above, one cytokine in particular, IL-12, has been shown to demonstrate in vivo anti-tumor activity in a wide variety of murine tumor models. For example, mice treated intraperitoneally with IL-12 were found to exhibit anti-tumor and anti-metastatic activity against a variety of different mouse tumors (including melanomas, hepatic tumors and sarcomas) [40–42]. Conversely, IL-12 deficiency has been shown to promote photocarcinogenesis in mouse skin [43]. Interestingly the same group found that IL-12 deficiency was also associated with an enhanced resistance to the development of TPA-induced skin tumors in DMBA-initiated mouse skin, due to a reduction in the DMBA/TPA-induced inflammatory responses in the skin and tumors of these mice (possibly due to their lower expression levels of COX2 and pro-inflammatory cytokines) [44]. Thus the role of IL-12 in chemical carcinogenesis is not so clear. 4.1.2. Tumor necrosis factors Tumor necrosis factors (or the TNF-family) are a group of cytokine family members that can cause apoptosis. Nineteen cytokines have been identified as part of the TNF family, however, monocytederived tumor necrosis factor-alpha (TNF-α) is the best-known member of this class, and sometimes referred to when the term “tumor necrosis factor” is used. TNF-α is a major mediator of inflammation, however its role in tumor growth and dissemination is context-dependent, as although TNF-α was originally identified by its ability to induce the necrosis of transplanted tumors in mice [45], subsequent clinical trials have shown that local administration of TNF-α at high concentrations produces a powerful anti-cancer activity by causing selective damage and obliteration of intratumoral blood vessels, yet there is also evidence that TNF-α possesses mitogenic activity and can enhance the ability of tumor cells to metastasize (reviewed in Ref. [46]). For example, mice deficient in TNF-α show a dramatic reduction (up to 90%) and delayed onset of chemical-induced skin tumors [47–49] and mice transplanted with a breast carcinoma cell line showed reduced tumor growth when dosed weekly with an anti-murine TNF-α antibody (cV1q), compared to transplanted mice dosed with vehicle alone [49]. Thus it has been proposed that the effects of TNF-α on tumor development are context dependent: high-dose local delivery of TNF-α can cause tumor regression, yet sustained production of endogenous TNF-α in the tumor microenvironment can enhance cancer development and metastasis (reviewed in Ref. [50]). Interestingly, the tumor resistance phenotype associated with loss of TNF-α can be partially recapitulated by loss of one of its major down-stream targets, monocyte chemoattractant protein-1 (MCP-1), as mice carrying a knockout allele of Mcf1 showed a ~50% reduction in chemical-induced skin tumors compared with wildtype mice [48]. In addition to being a key mediator of inflammation, TNF-α elicits pleiotropic effects in a wide range of cells by binding and activating two cell-surface receptors, specifically TNF receptors 1 and 2 (TNFR1 and R2). Similar to the conflicting reports of TNF-α in cancer resistance, the role of TNFR1 in cancer resistance is context dependent. For example, one study found that mice carrying a knockout Tnfr1 allele showed a reduced incidence of carcinogenic dietinduced liver tumorigenesis (interestingly the same protective effect was not achieved in Tnfr2 null mice) [51]. In contrast, in colitisassociated carcinogenesis TNFR1 functions as a tumor suppressor, with histological inflammation scores being higher and neoplastic lesions occurring more frequently and earlier in Tnfr1 null mice subjected to chemical-induced colitis, compared to controls [52]. Nevertheless, taken together, these studies confirm that ablation of the TNF signaling pathway can confer a cancer resistant phenotype. 4.1.3. CTLA-4 Activation of T lymphocytes is thought to require at least two signals, one delivered by the T-cell receptor (TCR) complex after antigen recognition, and one provided on the engagement of co-stimulatory receptors, such as CD28 (which binds CD80 and CD86 on APCs). However, other molecules can bind CD80/86, such as cytotoxic Tlymphocyte antigen 4 (CTLA-4; CD152). In contrast to co-ligation of the TCR and CD28, which results in T-cell activation, cytokine production, proliferation and differentiation, co-ligation of the TCR and L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 Table 2 Genetically engineered mice displaying cancer resistance phenotypes. The type of resistance includes the development of spontaneous or carcinogen-induced tumors (such as those arising naturally or after exposure to chemicals or irradiation), the development of genetically-induced tumors (by breeding to mice that carry genetic mutations predisposing them to the development of tumors) or the development of non-autochthonous tumors after transplantation (either intravenously, subcutaneously, intradermally, intracranially or intraperitoneally) with human or mouse tumor cell lines (with the resistance to tumor formation arising from over-expression or knock-down of a specific gene in the tumor cells or administration of a specific inhibitor). Genetic manipulation T-cell signaling Loss of IL-12 Loss of Tnfα Loss of Mcp-1 Loss of Tnfr1 Expression of CTLA4-Ig Inflammation Expression of Pla2g2a Loss of Pla2g4 Loss of Ptgs-2 Loss of Ptgs-1 Loss of Nos-2 Loss of Ikkβ Expression of Ikkα Expression of fat-1 Cell proliferation and death Expression of p53 Expression of Ink4a/Arf Expression of Pten Loss of c-Jun Loss of c-Fos Expression of Tgfβ Loss of Smad3 Expression of Bmp-4 Expression of Bmp-6 Expression of Bcl2 Loss of Bcl-xL Loss of Ptp1b Expression of Rassf3 Expression of PKCδ Expression of Gata4 Expression of Gata6 Expression of Par-4 Genomic stability/integrity Loss of Ppm1d Loss of Parp-1 Loss of Dnmt1 Loss of Terc Expression of Mgmt Resistance to spontaneous or carcinogeninduced tumors Resistance to geneticallyinduced tumors [44] [47,48] [48] [51] [55] [68] [77] [77] [49] [62,63,67] [69,70] [74,75] [75] [86,87] [91] [93] [97,98] [76,78] [99] [104–106] [110,111] [115] [118] [120] [124,127] [129] [134] [133] [139,140] [137] [119] [125] [126] [138] [148] [152] [155] [166] [166] [174] [181] [170] [182] [62,208–211] [214–219] [202–205,244] [213,214] [193–201] Angiogenesis Loss of Fgfbp Adhesion Loss of EphA2 Resistance to transplanted tumor cell lines [161] [159] [164,165,167] [175] [182,188] [220–223] [232,233] [228,229] [226,227] CTLA-4 results in cell-cycle arrest and termination of T-cell activation (reviewed in Ref. [53]). A soluble recombinant form of CTLA-4, CTLA41g, has been used as a competitive inhibitor of CD28 activation and can inhibit T cell-dependent immune responses, including induction of anti-tumoral immune responses, rejection of transplanted organs, and autoimmune responses [54]. In agreement with this, a CTLA-4Ig transgenic mouse model (K14-CTLA-4Ig), that expressed high amounts of CTLA-4Ig in the skin and systemic circulation, exhibited significant suppression of skin tumor formation after being chronically irradiated with UV [55]. Blockade of CD80/CD86CD28-CTLA-4 interactions in these mice significantly altered the Th1/Th2 balance in favor of Th1-mediated immune responses, and 317 thus reversed the chronic UV radiation-induced tendency to generate Th2 rather than Th1 immune responses [56]. 4.2. Inflammation 4.2.1. Phospholipase A2 enzymes Phospholipase A2 (PLA2) enzymes hydrolyze the fatty acid from the sn-2 position of membrane phospholipids. The released polyunsaturated fatty acids can be subsequently metabolized into eicosanoids (such as prostaglandins and leukotrienes) that are involved in numerous homeostatic biological functions [57]. The PLA2 superfamily comprises four main types including the secreted (s)PLA2, cytosolic (c)PLA2, calcium-independent (i)PLA2, and lipoprotein-associated (Lp)PLA2 (reviewed in Ref. [58]). The mouse sPLA2 group IIA gene Pla2g2a has been proposed to play a role in anti-bacterial defense, inflammation and eicosanoid generation, clearance of apoptotic cells, and the Wnt signaling pathway (reviewed in Ref. [59]). The evidence for Pla2g2a being involved in cancer resistance is linked to the ‘Min’ mouse strain. Min (Apc +/Min) mice carry a point mutation that disrupts the mouse homolog of the human familial polyposis gene (APC) and they develop intestinal neoplasms [60]. Multiplicity of these neoplasms is affected by genetic background, and a strong modifier locus, Mom1 (‘modifier of Min-1’) that suppressed Min-induced tumorigenesis was found in the AKR strain [61]. The Mom-1 region was subsequently mapped to mouse chromosome 4 and found to contain Pla2g2a and a locus distal to D4Mit64 [62], and Pla2g2a transgenic mice show a reduction in Mininduced intestinal tumors [62,63]. Interestingly, a second modifier of the Min phenotype, Mom-2, was serendipitously detected while further investigating Mom-1 in additional inbred strains [64] and this spontaneously arising mutation, specifically a 4 base-pair duplication in the coding sequence of Atp5a1 (an ATP synthase component), conferred a dominant, resistant phenotype with reduced polyp multiplicity [65]. Dove and co-workers were able to refine the locus of an additional modifier of the Min phenotype, Mom-7, to within the first 7.4 Mb of chromosome 18, and there are possibly five segregating alleles that likely to act by modulating net tumor growth or initiation [66]. However, the mechanism by which Pla2g2 performs its role in tumor resistance is still unclear. There is evidence for it being an Apc-independent role as expression of a Pla2g2a transgene reduced tumorigenesis 90–100% in the large intestine of mice carrying a knockout Muc2 allele (Muc2 −/− mice develop of adenocarcinomas in the small and large intestine) [67] and provided protection against carcinogen-induced duodenal and colon tumors (the tumors demonstrated upregulation of β-catenin which is indicative of involvement of the Wnt signaling pathway) [68]. Indeed, several models have been proposed to illustrate its putative biological effects on tumor development (reviewed in Ref. [59]). The cPLA2 group IV family member PLA2G4 catalyzes the hydrolysis of membrane phospholipids to release arachidonic acid, which is subsequently metabolized into prostaglandins, which in turn regulate many key intracellular pathways, in particular the inflammatory response. Thus inhibition of PLA2G4 would decrease the amount of arachidonic acid available to be metabolized into prostaglandins and as a result impair tumorigenesis. In agreement with this, Hong and colleagues found that Min mice carrying a knockout allele of Pla2g4 showed decreased prostaglandin production and an 83% reduction in tumors of the small intestine compared with littermates (Pla2g4 +/− and Pla2g4 +/+ mice on a Min background) [69]. Similarly, mice carrying a truncated Apc allele (Apc +/Δ716 mice) and a knockout allele of Pla2g4 showed a significantly reduced size of small intestinal polyps (although the number of polyps remained unchanged) [70]. 4.2.2. Cyclooxygenase enzymes Cyclooxygenase (COX) enzymes are responsible for the conversion of arachidonic acid to prostaglandins, which in turn can play a 318 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 Table 3 Transplant and xenograft mouse models have demonstrated that secretion of Th1-type cytokines or chemokines from tumors produces anti-tumor effects. Cytokine/ chemokine Immune system role Transduced cells Phenotype of the recipient mice IL-12 Stimulates production of IFN-γ in Th cells, thereby promoting the Th1 profile. Murine colon carcinoma cell line (colon 26) BALB/c mice showed an initial tumor take followed by a complete tumor regression and subsequent acquired tumor-specific protective immunity [249]. Nude or SCID mice produced tumors, however they survived longer than those injected with parental cells [250]. BALB/c mice showed an initial tumor take followed by complete regression and subsequent acquired tumor-specific protective immunity. Nude or SCID mice produced tumors but their overall survival was significantly longer than mice inoculated with parental cells [251]. Nude and SCID mice produced tumors, however nude mice survived longer than those injected with parental cells, unlike SCID mice [250]. BALB/c mice showed a significantly inhibited tumor growth (and in other experiments, direct injection of recombinant IL-18 into established tumors also inhibited tumor growth) [252]. IL-15 Secreted by mononuclear phagocytes (and some other cells) following viral infection and regulates T and NK cell activation and proliferation. Human pancreatic cancer cell line (AsPC-1) Murine colon carcinoma cell line (colon 26) Human pancreatic cancer cell line (AsPC-1) IL-18 IL-21 IL-23 IL-27 Produced by many cell types, it is a potent inducer of IFN-γ by T cells and NK cells and is synergistic in this function with IL-12. It can drive differentiation of CD4+ T cells to the Th1 phenotype (and can also promote Th2 responses under some conditions). Secreted primarily from CD4+ T cells, it induces proliferation of NK and activated T cells and increases the lytic functions of NK and cytotoxic T cells. Secreted from activated DC, and enhances proliferation of memory T cells and production of IFN-γ from activated T cells. It is structurally related to IL-12, thus shares its biological activities in facilitating Th1 responses. Produced primarily from activated DC, it is involved in an early phase of Th1 differentiation. It also synergizes with IL-12 in IFN-γ production from naive T and NK cells. Murine prostate carcinoma cell line (RM1) Human oesophageal tumor cell line (T.Tn) Human pancreatic cancer cell line (AsPC-1) Murine colon carcinoma cell line (colon 26) Human oesophageal tumor cell line (T.Tn) Murine melanoma cell line (B16-F1) Murine colon carcinoma cell line (colon 26) Murine neuroblastoma cell line (TBJ) CCL19 CCL21 CXCL4 (PF-4) Produced by a subset of DC and possibly by other non-lymphoid cells, in T-cell areas of lymphoid tissue. Its ability to chemoattract T cells, B cells, DC, macrophage progenitor cells and NK cells, is mediated through the CCR7 receptor. Constitutively expressed by high endothelial venules in lymph nodes and Peyer's patches, lymphatic vessels and stromal cells in spleen and appendix. It binds to the CCR7 receptor and is chemotactic for mature DC, naive and memory T cells. Along with CCL19, it is required for normal lymphoid tissue organization that is essential for effective T cell-DC interactions. Released from alpha-granules of activated platelets during platelet aggregation. It interacts with the CXCR3B receptor and is chemotactic for neutrophils, fibroblasts and monocytes. Murine breast cancer cell line (C3L5) Murine DC cells Human head and neck squamous carcinoma cell line (KB) Murine Lewis lung carcinoma cell line (LLH) Human myeloma cell lines (LP-1, U266, RPMI8226) Nude mice developed small tumors that regressed spontaneously thereafter, in contrast to parental cells [253]. Nude mice developed tumors, but at a retarded growth rate relative to parental cells [39]. BALB/c mice developed tumors, followed by a complete tumor regression and subsequent acquired tumor-specific protective immunity. BALB/c mice given tail vein injection showed significantly reduced number of lung metastasis relative to parental cells. Nude mice developed tumors, but at a retarded growth rate relative to parental cells [254,255]. Nude mice developed small tumors which regressed spontaneously thereafter, in contrast to parental cells [253]. C57BL/6J mice given tail vein injection showed significantly reduced number of lung metastasis relative to parental cells [255]. BALB/c mice rejected the tumors and subsequently acquired tumor-specific protective immunity. Nude mice developed tumors, but the growth was retarded compared to that of parental cells, however the survival of SCID mice was not different from that of the nude mice inoculated with parental cells [38]. A/J mice developed small tumors, followed by a complete tumor regression, with most showing subsequent acquired tumor-specific protective immunity. Tail vein injection showed significantly reduced number of liver metastasis relative to parental cells [256]. Only 10% of C3H/HeN mice injected developed tumors (compared with 100% of mice injected with a transduced control) and subsequently acquired partial tumor-specific protective immunity [257]. BALB/c mice subcutaneously administered the murine lung tumor cell line, L1C2, and 5-days later received an intratumoral injection DC transduced with CCL21, showed complete tumor eradication (60% of mice compared to 12% of mice that received unmodified DC cells) [258]. Transgenic mice (CC-10 TAg) that develop pulmonary adenocarcinomas by 4 months, showed a marked reduction in tumor burden with extensive mononuclear cell infiltration of the tumors after a single intra-tracheal administration of CCL21 gene-modified DC [259]. Nude mice showed an inhibition of solid tumors through an anti-angiogenic action [260]. Nude mice given tail vein injection showed significantly reduced number of lung metastasis [261]. SCID mice showed significant reduction in microvessel densities in human myeloma xenografts and markedly reduced the tumor volume, which significantly extended their overall survival [262]. Data in this table are derived from cancer cell lines that are retrovirally transduced with specific a cytokine or chemokine gene and then transplanted (intraperitoneally or subcutaneously, unless stated otherwise) into recipient mice. However, it should be noted that many of these cytokines and chemokines have also been shown to have an antitumoral effect when administered as a recombinant protein. For example, administration of recombinant CXCL4 effectively suppressed the growth and lung metastases of a murine melanoma cell line (B16-F10) in syngeneic C57BL/6J hosts and prevented the growth of primary tumors of B16-F10 and a human colon carcinoma cell line (HCT-116) in semi-syngeneic CByB6F1/J mice [245,246]. In addition, BALB/c mice subcutaneously administered a murine lung tumor cell line (L1C2), and 5-days later receiving an intra-tumoral injection of recombinant CCL19, showed significant systemic reduction in tumor volumes [247], and transgenic mice that develop pulmonary adenocarcinomas by 4 months, showed a marked reduction in tumor burden with extensive mononuclear cell infiltration of the tumors after a single intra-nodal administration of recombinant CCL21 [248]. A/J, BALB/c, C3H/HeN and C57BL/6J are immunocompetent mice, whereas nude and SCID mice are immunocompromised. Abbreviations: DC, dendritic cells, DC; IFN-γ, interferon-γ; IL, interleukin; SCID, severe combined immunodeficient; Th, helper T cell. L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 role in the inflammatory response. COX exists in two main isoforms, COX-1, which is constitutive and responsible for generation of eicosanoids for “housekeeping functions”, and COX-2, the inducible isoform, which contributes eicosanoids involved in a variety of growth and inflammatory events [71,72]. Many malignant cells (particularly in breast and colorectal cancers) show over-expression of COX-2, while COX-1 expression remains unaltered [72], and non-steroidal anti-inflammatory drugs, which are known to inhibit COX enzymes, have shown to lower the mortality rate from cancers in the gastrointestinal tract [73]. Studies in the mouse have confirmed the approach of ablating COX-2 as an approach to protect against cancer. For example, mice carrying a knockout allele of Cox-2 (Ptgs-2 −/−) on a mutant Apc background (either Min or Δ716 mice) showed a dramatically reduced number and size of intestinal polyps [74,75]. Loss of COX-2 has also been shown to provide a protective effect in other tumor types, with Ptgs2 −/− embryonic stem cells showing a dramatic reduction in ability to form teratocarcinomas when injected into syngeneic mice [76], and Ptgs2 −/− mice showing a 75% reduction in chemically-induced skin papilloma formation [77]. Nude mice that received Celecoxib (a COX-2 inhibitor) showed reduced growth and lymphangiogenesis of human mammary cancer xenografts as compared with controls [78]. Conversely, transgenic COX-2 overexpression sensitizes mouse skin for carcinogenesis, with keratin 5 promoter-driven overexpression of COX-2 in basal keratinocytes contributing to skin-tumor promotion and progression by establishing an “auto-promoted” skin phenotype, i.e., the initiating dose of DMBA was sufficient to induce skin carcinogenesis in COX-2 transgenic mice [79]. However, in contrast to these reports, one study found that over-expression of COX-2 provided a protective effect against chemically-induced skin papilloma formation with transgenic mice over-expressing COX-2 under the control of the human keratin-14 promoter (K14.COX-2 mice) developing skin tumors at a much lower frequency than wildtype littermates [80]. Further analysis of this model to reconcile these findings revealed that the effect of COX-2 over-expression on skin carcinogenesis is context dependent, with the mechanism of this resistance restricted to TPA promotion, as K14.COX-2 mice developed more tumors than wildtype mice when anthralin was used as the tumor promoter or when they were treated only with DMBA [81]. Although the role of the COX enzymes in tumorigenesis has predominantly headlined COX-2, there is some evidence that COX-1 also has a role to play. For example, studies using mice carrying a knockout allele of Cox-1 (Ptgs-1 −/−) found a 75% reduction in DMBA/TPA-induced skin papilloma formation [77] and ~ 80% reduction in polyp formation on a Min background [75]. Interestingly, the COX-1 and −2 inhibitor, piroxicam, reduced the growth of canine mammary cancer xenografts in nude mice unlike mice treated with vehicle or the selective inhibitor of COX-2, deracoxib [82]. Thus at least in some tumor types, there are comparable contributions of both COX isoforms in the development of cancer. 4.2.3. Nitric oxide synthases Nitric oxide (NO) is a short-life molecule produced by the nitric oxide synthase (NOS) enzyme, in a reaction that converts arginine and oxygen into citrulline and NO. There are three isoforms of the enzyme: neuronal NOS (nNOS/NOS-1), inducible NOS (iNOS/NOS2), and endothelial NOS (eNOS/NOS-3). NO plays a role in a multitude of diseases, including cancer, myocardial and central nervous system pathologies, and inflammation (reviewed in Ref. [83]). NOS-2 is cytokine-inducible, calcium/calmodulin-independent, expressed in essentially every cell type and can locally generate high quantities of NO at micromolar range for prolonged periods of time [84]. Numerous reports suggest that NO can have tumor-promoting effects, and as such NOS-2 expression has been reported as having tumor-promoting effects (reviewed in Ref. [85]). In agreement with this, mice carrying knockout Nos-2 alleles on a Min background show significantly 319 reduced intestinal adenoma formation [86,87]. In contrast, several clinical and experimental studies indicate that the presence of NO in tumor microenvironment is detrimental to tumor cell survival and metastasis, and that NOS-2 expression can have anti-tumor effects (reviewed in Ref. [85]). For example, one study found that mice carrying a knockout Nos-2 allele on a Min background developed significantly more intestinal adenomas than Apc Min/+, Nos2 +/+ littermates [88]. Interestingly, another study found no difference in ulcerative colitis (UC)-associated colo-rectal cancer development in wildtype and Nos-2 −/− mice, and suggests that in the absence of NOS-2, other factors such as NOS-3, may play a role in nitrosative stress and UC-associated carcinogenesis [89]. It has been suggested that the dichotomy of NOS-2 expression in cancer can be reconciled by consideration of the concentrations of NO involved, the temporalspatial mode of NO action, intracellular targets, cellular redox state and the timing of an apoptotic stimulus (reviewed in Ref. [85]). However, for now, the cancer resistant phenotype of the Nos-2 null mice needs further investigation. 4.2.4. IkappaB kinase complex The IkappaB (IκB) kinase (IKK) complex includes the catalytic subunits IKKα and IKKβ and the scaffold protein NEMO (also known as IKKγ). The IKK has a crucial role in the activation of the transcription factor nuclear factor kappa B (NF-κB) by phosphorylating the inhibitory molecule IκBα, which triggers the subsequent polyubiquitylation and degradation of IκBα through the proteasome, allowing the derepressed NF-κB to bind target genes involved in cell proliferation, survival and immunity. More recently, the IKK has also been identified as having key roles in other processes ranging from allergy to cancer through NF-κB-independent pathways (reviewed in Ref. [90]). Although IKKα and IKKβ share considerable sequence identity, it is IKKβ that usually serves the more critical function in the activation of classical NF-κB signaling. Mice that specifically lack IKKβ in their intestinal epithelial cells (villin-Cre/Ikkβ F/Δ mice) show a dramatic decrease of AOM-DSS-induced 75% in tumor incidence, and mice that specifically lack IKKβ in the myeloid lineage (LysM-Cre/ Ikkβ F/F mice) show an almost 50% reduction in tumor counts, relative to the Ikkβ F/Δ controls [91]. Thus IKKβ acts both within enterocytes, which eventually give rise to the transformed component of the tumor, and within myeloid cells, which influence tumor growth; in each cell type IKKβ contributes to tumor promotion through a different mechanism, specifically by suppressing apoptosis in epithelial cells and regulating expression of inflammatory mediators in myeloid cells [91]. In contrast, IKKα has been shown to function as a molecular switch that controls epidermal differentiation, and a reduction of IKKα expression promotes skin tumor development, with IKKα hemizygote (IKKα +/−) mice developing 2- and 11-times more papillomas and carcinomas, respectively, than wildtype mice 3 [92]. The same group also showed that mice over-expressing human IKKα in the suprabasal compartment of the epidermis, which results in increased epidermal differentiation and reduced keratinocyte proliferation, developed significantly fewer chemically-induced squamous cell carcinomas and metastases than wildtype mice [93]. 4.2.5. Polyunsaturated fatty acids Polyunsaturated fatty acids (PUFA) belong to two families: n-6 and n-3 PUFAs. Mammals cannot naturally produce n-3 PUFA from the more abundant n-6 PUFA, so they must rely on a dietary supply. n-3 PUFA have been implicated in inflammation dampening and there is evidence to suggest that they are potentially protective 3 IKKα-deficient (Ikkα−/−) mice could not be used as they show impaired formation of the skin, resulting in death soon after birth. 320 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 against several cancers, such as colon or breast cancers, mainly on the promotion and progression stages, whereas n-6 PUFA tend to favor cancer development [94]. The fat-1 gene of Caenorhabditis elegans encodes an n-3 fatty-acid desaturase enzyme that converts n-6 to n-3 PUFA and transgenic mice carrying fat-1 gene can convert n-6 to n-3 PUFA, resulting in an abundance of n-3 and a reduction in n-6 PUFA in these mice [95]. The increased levels of n-3 PUFA in these mice confer protection to acute DSS-induced colitis [96] and the same group showed that these mice have a lower incidence and growth rate of DSS/AOM-induced colon tumors, via an n-3 PUFA-mediated dampening of inflammation and NF-κB activity [97]. More recently, the group found that these mice were also more resistant to liver cancer, with the development of fewer DEN-induced liver tumors relative to controls, due to an n-3 PUFA-derived 18-HEPE and 17-HDHA-mediated down-regulation of TNF-α [98]. Transgenic fat-1 mice implanted with mouse melanoma B16 cells also show a reduced incidence of tumor formation and growth rate relative to controls, mediated by the n-3 PUFA metabolite prostaglandin E3-induced activation of PTEN pathway [99]. [104] (Fig. 2). Similarly, p44 mice showed a very low incidence of spontaneous tumors, and showed signs of premature aging [105] (Fig. 2). Interestingly, one p53 mouse model was able to uncouple the cancer resistance phenotype from the premature aging phenotype by proper regulation of full-length p53 expression. “Super-p53” mice showed significantly increased resistance to the development of spontaneous and chemically-induced tumors compared to wildtype mice, and did not show any signs of premature aging — presumably as the p53 is not constitutively active, but rather activated only in response to the appropriate stimuli, where it can respond in an enhanced manner (Fig. 2) [106]. Paradoxically, one study found a tumor inhibitory effect of p53 loss in transgenic mice expressing epidermal-targeted Ras, Fos, or human TGF-α [107]. The authors proposed that the paradoxic block in papillomatogenesis in the p53 −/− background may occur by mechanisms that attempt to maintain epidermal homeostasis and could result from upregulation of additional p53 homologs, such as p73 and p63 (which exhibit some functions similar to p53 and are able to bind to consensus p53-binding sites of known transcriptional targets) [107]. 4.3. Cell growth/death 4.3.2. INK4a/ARF/INK4b locus The INK4a/ARF/INK4b locus (also known as CDKN2a and CDKN2b) is a small 35 kb region on human chromosome 9p21 that contains three related genes: ARF (also known as p19 ARF or p14 ARF), p15 INK4b, and p16 INK4a, that each encode distinct tumor suppressor proteins (reviewed in Ref. [108]). The tumor suppressor activities of p15 INK4b or p16 INK4a are related to their ability to inhibit the cell cycle, specifically maintaining Rb-family proteins in a hypophosphorylated state, which promotes binding E2F to effect a G1 cell-cycle arrest, whereas the tumor suppressor activity of ARF is largely ascribed to its ability to regulate p53 in response to aberrant growth or oncogenic stresses such as c-MYC activation (reviewed in Ref. [109]). Knockout studies of mice deficient for either of these genes have revealed that all three strains are more prone to spontaneous cancers than wildtype littermates whereas overexpression of the Ink4a/Arf/Ink4b locus confers a degree of cancer resistance (reviewed in Ref. [109]). From the same group that generated the "super-p53" mice (see above), mice that carry an allele of the Ink4a/Arf/Ink4b locus on a bacterial artificial chromosome and display modest overexpression of p16 INK4a, p15 INK4b, and Arf were generated (“super-Ink4/Arf” mice)[110]. “Super-Ink4a/Arf” mice treated with carcinogens (to induce skin papillomas, fibrosarcomas, plasmacytomas, and lung adenomas) are 4.3.1. p53 The tumor suppressor gene p53 is a major genome guardian molecule and responds to a diverse array of stresses that include DNA damage and aberrant oncogene signaling. Upon activation, p53 prevents the emergence of cancer cells by initiating cell cycle arrest, senescence, or apoptosis [100]. The fact that half of all human cancers display loss-of-function mutations or deletions in the p53 gene is a strong argument for the importance of p53 anti-cancer function [101]. Although early attempts to generate genetically engineered mouse models over-expressing p53 (either globally or in a tissuespecific manner) have not always been successful due to issues of embryonic lethality or premature degeneration of the targeted tissue (reviewed in Ref. [102]), models using only moderately elevated (or correctly regulated) levels of p53 expression have had success in showing that boosting p53 levels confers cancer resistance (reviewed in Ref. [103]). Heterozygous p53 mice expressing the ‘m’ allele (p53+/m) displayed a phenotype consistent with ‘activated p53’, showing enhanced resistance to the generation of spontaneous tumors compared with control littermates (p53+/+ and p53+/−), although they also displayed a multitude of phenotypes consistent with premature aging p53+/m mouse p44 mouse Super-p53 mouse p53 p53 p53 p53 m p53 p53 p44 p53 Viable More resistant to cancer Premature aging Viable More resistant to cancer Premature aging Viable More resistant to cancer No change in aging Fig. 2. Examples of genetically engineered p53 mutant mice showing cancer resistance phenotypes. Wildtype (endogenous) p53 alleles are shown in gray, and modified or exogenous (transgenic) p53 alleles are shown in red. p53m mice carry one p53 allele that contains a deletion of the 5′ UTR and exons 1–6 (constituting two-thirds of the protein), so produces only a C-terminal fragment that marginally enhances the transactivation and DNA binding activity of the wildtype protein [104]. Transgenic p44 mice carry a stably integrated copy of the p44 gene (a ‘short form’ of the p53 allele in which translation initiates at codon 41 in exon 4 and produces a 44 kDa protein that is unable to transactivate target genes such as Mdm2 and p21/Cip1/Waf1 [105]). Transgenic Super-p53 mice carry a stably integrated copy of the p53 gene in the form of a large genomic transgene (contained in a bacterial artificial chromosome) that contains > 100 kb of the surrounding genomic sequence of the gene [106]. L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 more resistant to tumor development than their wildtype littermates, and also display a lower incidence of spontaneous tumors [110]. In addition, when compared with “super p53” or “super Ink4a/Arf” mice, “super Arf/p53” double transgenic mice showed a delay in the latency of chemically-induced fibrosarcomas and papillomas, as well as a significantly diminished incidence of sporadic cancer in aged mice [111]. 4.3.3. Phosphatase and tensin homologue The tumor suppressor gene product of the phosphatase and tensin homolog (PTEN) gene is a lipid phosphatase, which reduces the cellular levels of phosphatidylinositol-3-phosphate (PI3P) by antagonizing the activity of phosphoinositol-3 kinase (PI3K). Loss of PTEN activity leads to the accumulation of cytosolic PI3P and activation of the AKT pathway, which results in increased cell growth, survival, invasiveness, and metabolism, whereas over-expression of PTEN in cell culture down-regulates cyclin D1 expression and results in cell cycle arrest [112]. PTEN was the first phosphatase identified to be frequently mutated or to show somatic deletions in various human cancers and Pten-deficient (Pten +/−) mice develop hyperplastic or neoplastic changes in many organs at an early age [113]. Over-expression of PTEN specifically in the mammary gland of mice results in a marked decrease in mammary epithelial cell proliferation and an increase in epithelial cell apoptosis [114], and was later found to confer a resistance to mammary tumors. Specifically, over-expression of the proto-oncogene Wnt-1 in mammary epithelium leads to mammary hyperplasia and subsequently focal mammary tumors, however, mice that ectopically express PTEN and Wnt-1 in mammary epithelium showed a reduction in the onset (5.9 to 7.7 months), growth rate (time to grow from 0.5 to 1 cm was 8.4 to 17.7 days) and number (11% reduction) of Wnt-1-induced tumors within a 12-month period [115]. Earlier this year, a “Super-PTEN” mouse was generated (similar to the generation of “Super-p53” and “Super-Ink4a/Arf” mice above) which carries a large genomic fragment encompassing the entire Pten locus and shows a cancer resistance phenotype, specifically reduced susceptibility to chemically-induced fibrosarcomas [116]. 4.3.4. Activator protein 1 Activator protein 1 (AP1) functions in almost all areas of cellular behavior and is activated in response to a variety of extracellular signals from cytokines and growth factors to stress and inflammation. The expansive transcriptional repertoire mediated by AP1 complexes is propagated by the diverse array of homo- or hetero-dimeric combinations formed by members of the Jun, Atf, Fos, Fra and Maf transcription factor families which are often deregulated by oncoprotein signaling in malignant cellular transformation (reviewed in Ref. [117]). c-Jun levels and N-terminal phosphorylation (which is crucial for its activation) are cell cycle regulated, and over-expression of Jun promotes cell growth in many cell lines whereas mouse fibroblasts lacking c-Jun exhibit severely impaired proliferation (reviewed in Ref. [117]). This inhibition of proliferation leads to a cancer resistance phenotype as transgenic mice expressing TAM67 (a dominantnegative form of c-Jun that lacks the N-terminal transactivation domain) under control of the human keratin-14 promoter show a dramatic inhibition of DMBA/TPA-induced skin papillomas [118]. In addition, expression of TAM67 in non-small cell lung cancer NCIH1299 cells reduced the growth of established xenograft tumors from these cells in nude mice [119]. A similar benefit was seen with loss of functional expression of the AP1 subunit c-fos, as c-fos −/− mice carrying a v-H-ras transgene developed TPA-induced skin papillomas with similar kinetics and incidence to wildtype mice, however, whereas wildtype papillomas progressed to malignant tumors, c-fosdeficient tumors failed to undergo malignant conversion [120]. In addition, the life span of 60% of BALB/c mice dosed with a DNA vaccine against murine transcription factor Fos-related antigen 1 (Fra-1), was 321 tripled in the absence of detectable tumor growth after lethal administration of murine breast carcinoma D2F2 cells [121]. 4.3.5. Transforming growth factor-β Transforming growth factor-β (TGF-β) initiates downstream signaling events by activation of transmembrane serine-threonine kinase receptors, TGF-β type I and II receptors (TGF-βRI and TGFβRII) and intracellular Smad effectors, regulating numerous epithelial cell processes. TGF-β plays a crucial role in cancer initiation and progression through tumor cell autonomous signaling and interactions with tumor microenvironment. The complexity associated with TGF-β regulation of cell behavior was evident in early experiments that demonstrated TGF-β-mediated growth inhibition in culture conditions that previously stimulated progressive anchorage independent growth, and primary keratinocytes responding to TGF-β stimulation with arrest of the cell cycle in G1 while a number of other cancer cell lines were able to evade this response. Thus TGF-β can both induce growth inhibition and support anchorage independent growth, however the responses are cell type and context dependent (reviewed in Ref. [122]). More recently, it has been found that TGF-β signaling acts as a suppressor of epithelial cell tumorigenesis at early stages, but promotes tumor progression by enhancing migration, invasion, and survival of the tumor cells during the later stages (reviewed in Ref. [123]). Consistent with a growth inhibitory effect arising from TGF-β expression, studies in mice have shown that transgenic mice over-expressing TGF-β in certain cell types can result in a cancer resistance phenotype. For example, transgenics overexpressing TGF-β in mammary tissue do not develop mammary tumors, are resistant to DMBA-induced mammary tumor formation [124], and show reduced MMTV-induced mammary tumor formation [125]. Highly malignant K-ras-transformed thyroid cells showed a decrease in tumorigenicity in nude mice in vivo when they were transfected with the human TGF-βRII gene [126]. Interestingly, transgenic mice with keratinocyte-targeted TGF-β expression are more resistant than controls to DMBA/TPA-induced benign skin tumors, however, the malignant conversion rate was vastly increased in the transgenic mice compared to control mice [127]. Thus, the diversities of TGF-β signaling in tumors imply a need for caution to TGF-β-targeted strategies of tumor prevention and/or therapeutics. 4.3.6. Smad3 Smads are the central mediators converting signals from receptors for TGF-β superfamily members to the nucleus. Catalytically active TGF-β type I receptor (TGF-βRI) phosphorylates the C-terminal serine residues of receptor-activated Smads (R-Smads), which include Smad2 and Smad3. TGF-βRI and Ras-associated kinases differentially phosphorylate Smad2 and Smad3 to create three phosphorylated forms, and phosphorylated Smad2 and Smad3 rapidly oligomerize with Smad4 and translocate to the nucleus to regulate transcription of target genes. Reversible shifting of Smad3-mediated signaling between tumor suppression and oncogenesis in hyperactive Rasexpressing epithelial cells indicates that the TGF-βRI-dependent phosphorylated form of Smad3 transmits a tumor-suppressive TGFβ signal, whereas oncogenic features such as cell growth and invasion are promoted through the JNK-dependent phosphorylated Smad3 pathway (reviewed in Ref. [128]). Since Smad3 has been shown to exert both tumor-suppressive and -promoting roles in a contextdependent manner, the role of Smad3 in skin carcinogenesis was examined. Homozygous knockout Smad3 (Smad3 −/−) mice are resistant to TPA-induced epidermal hyperproliferation, show reduced DMBA/ TPA-induced papilloma formation comparison with wildtype mice, and do not develop squamous cell carcinomas [129]. TPA-induced Smad3−/− papillomas showed reduced expression levels of AP-1 family members and reduced leukocyte infiltration, particularly macrophages, in comparison with wildtype papillomas [129]. Interestingly a previous study found that Smad3 −/− mice showed accelerated 322 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 cutaneous wound healing compared with wildtype mice, characterized by an increased rate of re-epithelialization and reduced local infiltration of monocytes [130]. Thus loss of Smad3 may be beneficial for tumor resistance as it eliminates the ability of TGF-β to exert its potent chemotactic effect on macrophages and thus impairs the local inflammatory response. 4.3.7. Bone morphogenetic proteins The bone morphogenetic protein (BMP) family is part of the TGF-β superfamily. Engaging of BMPs to their cell surface receptors leads to activation of the receptor kinase activity, which phosphorylates Smad-1, -5 or ‐8, allowing formation of complexes with Smad4 and subsequent nuclear translocation and regulation of transcription of BMP target genes. BMP-Smad signaling regulates stem cell renewal, cell proliferation, differentiation, migration, and apoptosis. In addition, BMP-Smad signaling has been shown to play an important role in tumorigenesis, with increasing evidence indicating that in many tissues, BMP-Smad signaling has a tumor-suppressing activity and that BMPs can repress tumor growth (reviewed in Ref. [131]). For example, although transgenic mouse strains epidermally-expressing BMP-6 (under control of the keratin 10 promoter) show keratinocyte hyperproliferation as well as inflammation [132], they are resistant to DMBA/TPA-induced skin tumor formation [133]. The authors postulated that this was due to the increased apoptotic frequencies and downregulation of AP-1 constituents observed in the keratinocytes of these mice [133]. Similarly, transgenic mice epidermallyexpressing BMP-4 (under control of the cytokeratin 4 promoter) do not develop skin tumors after treatment with MNNG/TPA, compared to papillomas and SCCs developing in 50% of control mice [134]. The epidermis of these mice showed reduced mitotic indices and only minimal hyperproliferation, and inflammation in response to TPA [134]. 4.3.8. B-cell lymphoma 2 family B-cell lymphoma 2 (BCL2) proteins are important cell death regulators, whose main function is to control the release of cytochrome c from mitochondria in the intrinsic apoptotic pathway. The BCL2 protein family consists of both pro- and anti-apoptotic members, which all share sequence homology in their BCL2 homology (BH) domains. The pro-apoptotic proteins comprise the multidomain proteins BAX and BAK, and anti-apoptotic BCL2 proteins include BCL2, BCL-xL, BCL-w, MCL1, BCL-B and BCL2A1. Many of these proteins have been identified as important cellular oncogenes that not only promote tumorigenesis but also contribute to the resistance to chemotherapeutic drugs (reviewed in Ref. [135]). In addition, the importance of BCL2 proteins in cancer progression has recently been highlighted in a genome-wide screen identifying BCL-xL and MCL1 as highly amplified in cancer cells [136]. Bcl-xL is one of several anti-apoptotic proteins regulated by signal transducer and activator of transcription 3 (Stat3), which is required for chemically- and UVB-induced skin carcinogenesis. To investigate the functional role of Bcl-xL in skin carcinogenesis, skin-specific Bcl-xL-deficient mice were generated in which Bcl-xL expression was disrupted in the basal compartment of mouse epidermis using the bovine keratin 5 promoter to drive expression of Cre recombinase (K5.Cre x Bcl-xLfl/fl mice). These mice were more resistant than wildtype controls to skin tumor development with delayed onset and reduced number of tumors using either UVB or DMBA/TPA [137]. Bcl-2, Mcl-1, and survivin protein levels were increased in unstimulated Bcl-xL-deficient epidermis and carcinogenesisinduced skin tumors, suggesting that Bcl-xL plays a role early in skin carcinogenesis through its anti-apoptotic functions to enhance survival of keratinocytes following DNA damage [137]. In agreement with this, studies using transgenic mice overexpressing Bcl-2 showed that they were protected against c-mycinduced liver tumors (by inhibiting a pre-tumoral phase characterized by increased proliferation and apoptosis) [138], DEN-induced liver carcinogenesis (by delaying the growth of proliferative foci at the early stages of carcinogenesis and inhibiting cell proliferation in these foci) [139] and chronic UVB-induced tumors (as they developed tumors much later and at a significantly lower frequency than controls) [140]. However, these findings are in conflict with studies which have reported Bcl-2 knockout (Bcl-2 −/−) mice show retarded oncogene-induced tumor development (transgenic mice carrying lung targeted expression of Raf oncogenes take longer to develop tumors when on a Bcl-2 −/− background) [141], and transfection of human gastric cancer SGC-7901 cells with Bcl-2 siRNA significantly suppresses their ability to grow when injected subcutaneously into nude mice (these cells resulted in smaller tumors compared to cells transfected with non-silencing siRNA) [142]. Thus it may be that Bcl-2 can play different roles in different tissues and contexts. Indeed, opposing findings have reported for BCL-2 expression in tumors, for example, loss of BCL-2 expression correlates with tumor recurrence in colorectal cancer [143], whereas BCL-2 expression correlates with metastatic potential in pancreatic cancer cell lines [144]. 4.3.9. Protein tyrosine phosphatase 1B Protein-tyrosine phosphatase 1B (PTP1B) is a non-transmembrane protein tyrosine phosphatase that has long been studied as a negative regulator of insulin and leptin signaling (reviewed in Ref. [145]). Ptp1b-null mice are lean, resistant to weight gain on a high-fat diet hypersensitive to insulin and show increased insulin sensitivity [146]. PTP1B is also a negative regulator of cell growth [145] and loss of Ptp1b in a p53-null background (Ptp1b −/−, Trp53 −/− mice) leads to an increased incidence of B-cell lymphoma through the regulation of B-cell development [147]. However, it has also long been known that expression levels of PTP1B are elevated in a number of human cancers, particularly breast and ovarian cancer [145]. More recently, PTP1B has been shown to be a positive regulator of the ErbB2induced mammary tumorigenesis in mice, as Ptp1b-deficient mice crossed with transgenic mice over-expressing activated mutants of ErbB2 in mammary epithelial cells (MMTV/neu mice) showed a significantly delayed onset of breast cancer, and treatment of MMTV/neu mice with a specific PTP1B inhibitor resulted in significant mammary tumor latency and resistance to lung metastasis, whereas transgenic over-expression of PTP1B induced breast tumors in the absence of exogenous ErbB2 [148,149]. However, it is important to note that Ptp1b deficiency does not offer universal protection against breast cancer, as breast tumors driven by the polyoma middle TAg were not affected by loss of Ptp1b [149]. Thus, PTP1B appears to have a selective, positive role in oncogenic signaling from ErbB2, however, it is not known if this will apply in other oncogenic or tissue settings. Thus more investigation of PTP1B in tumorigenesis is needed, specifically to ensure that there are no possible cancer-promoting effects of chronic PTP1B inhibition. 4.3.10. Rassf3 RAS proteins belong to a large superfamily of small GTPases and are signal transduction proteins that can interact with a wide array of effectors through the RAS binding domain (RBD) to stimulate diverse cytoplasmic signaling pathways. Although typically associated with loss of growth control and tumorigenic transformation (reviewed in Ref. [150]), there is increasing that RAS proteins have the ability to activate a variety of growth-inhibiting pathways including apoptosis and cell cycle arrest. For example, four members of the Ras association domain family (RASSF), RASSF1, RASSF2, RASSF4 and RASSF5 have been identified as RAS effectors and tumor suppressors involved in the pathways mediating RAS growth-inhibitory effects (reviewed in Ref. [151]). In a search of the gene(s) responsible for naturally acquired resistance to breast cancer tumorigenesis found in a small percentage of MMTV/neu transgenic mouse, comparative genetic profiling was used to screen alterations in gene expression in the mammary gland between resistant and susceptible mice from L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 this strain [Jacquemart et al., 2009]. One of the genes identified as being over-expressed in mammary glands of tumor-resistant MMTV/ neu mice was Rassf3, and a MMTV/neu-Rassf3 bi-transgenic mouse line showed delayed mammary tumor incidence compared to MMTV/neu littermates [152]. Thus Rassf3 influences mammary tumor incidence in MMTV/neu transgenic mice, although the molecular mechanism of growth-inhibition of Rassf3 and its particular role in MMTV/ neu tumor initiation and progression needs further investigation. 4.3.11. Serine/threonine protein kinases The serine/threonine protein kinase C (PKC) family was first identified as intracellular receptor(s) for the tumor promoting agent phorbol esters. At present ten PKC isozymes have been discovered and are classified in three subfamilies according to the functional domain composition: classic/conventional (PKC-α, PKC-βI, PKC-βII and PKC-γ), novel (PKC-δ, PKC-ε, PKC-η and PKC-θ) and atypical (PKC-ζ and PKC-τ). PKC isozymes can redistribute inside the cell in response to apoptotic stimuli through intrinsic localization sequences or specific-scaffolding protein binding, and this spatial regulation seems to be functional to the phosphorylation of substrates present only in specific subcellular districts. PKC isozymes are involved in tumor progression and metastasis, with each isoform being unique in its contribution to cancer development and progression (reviewed in Ref. [153]). Of the novel PKC isoforms, PKC-ε confers a tumorigenic and metastatic invasiveness phenotype in nude mice [154], whereas PKC-δ seems to have possible tumor-inhibitory characteristics. For example, transgenic mice epidermally over-expressing PKC-δ (from the human keratin 14 promoter) showed a suppression of DMBA/ TPA-induced skin tumor formation (papillomas and carcinomas) relative to wildtype mice [155]. However, the tumor resistance phenotype associated with PKC-δ expression is tissue specific and does not extend to brain and pancreatic cancer, where it seems to have a more pro-oncogenic role. For example, human ductal carcinoma PANC1 cells over-expressing PKC-δ were more tumorigenic than control cells when subcutaneously administered to nude mice, and the mice also developed lung metastasis [156]. 4.3.12. GATA factors The GATA family of transcription factors consists of six members that have two highly conserved zinc-finger domains and recognize a consensus DNA-binding motif of (A/T)/GATA/(A/G). They regulate biological functions, including organogenesis, differentiation, proliferation and apoptosis, by activating or repressing transcription [157]. Thus GATA factors coordinate cellular maturation with proliferation arrest and cell survival, so a role in human cancers is not surprising (reviewed in Ref. [158]). Genetic alterations in GATA-6 have been found in human malignant astrocytoma specimens, and knockdown of Gata6 expression in transformed astrocytes leads to acceleration of tumorgenesis [159]. Conversely, re-expression of GATA6 in human glioblastoma U87 and U373 cells lacking GATA-6 expression reduced their tumorgenic growth in NOD-SCID mice, concomitant with inhibition of VEGF expression [159]. The same group identified GATA4 as being expressed in the embryonic and adult central nervous system and acting as a negative regulator of astrocyte proliferation and growth, via transcriptional induction of p15 INK4B, leading to attenuation of cyclin D1 [160]. Recently they showed that inducible re-expression of GATA4 in U87 cells suppresses their ability to form astrocytomas after intracranial injection in NOD-SCID mice [161]. 4.3.13. Prostate apoptosis response-4 The prostate apoptosis response-4 (par-4) gene was first identified by differential hybridization as an immediate early apoptotic gene upregulated in response to elevated intracellular Ca 2+ concentration in ionomycin-treated rat prostate cancer AT-3 cells [162]. Human Par-4 protein shares significant sequence similarity with its rat counterpart, and ubiquitous expression has also been observed 323 in nearly all tissues of mice, horses, pigs and cows. Par-4 is a cancer cell-selective proapoptotic protein that inhibits NF-κB-mediated cell survival mechanisms and is down-regulated in many cancers (reviewed in Ref. [163]). The apoptotic effect of Par-4 is evident in vivo as injection of a Par-4-expressing adenoviral construct into tumors generated by transplanted murine prostate PC-3 cells in SCID mice resulted in a drastic reduction in tumor volume within 3 weeks, due to an increase in apoptosis [164], and over-expression of Par-4 in tumors generated by xeno-transplanted human melanoma A375-C6 cells in SCID mice correlated with decreased tumor development and increased apoptosis [165]. Analysis of deletion mutants of Par-4 led to the identification of a unique core domain (spanning amino acids 137–195), which when over-expressed, induces apoptosis specifically in cancer cells, and therefore is called the ‘selective for apoptosis of cancer cells’ (SAC) domain. Consistent with the cancerselective apoptotic action of the SAC domain, GFP-tagged SAC transgenic mice show remarkable resistance toward the formation of spontaneous and oncogene-induced tumors [166], as well as nonautochthonous tumors (after transplant of mouse lung carcinoma LLC1 cells into their flanks) [167]. Recent findings indicate that Par4 is also secreted by cells and extracellular Par-4 induces cancer cell-specific apoptosis by interaction with the cell-surface receptor GRP78 [168]. In agreement with this, recombinant Par-4 or SAC protein intravenously administered to immunocompetent C57BL/6 mice significantly inhibited lung metastasis of LLC1 cells [167]. Thus both systemic (extracellular) and intracellular Par-4 may contribute to tumor resistance in SAC- and Par-4-transgenic mice. 4.4. Maintaining genomic stability/integrity 4.4.1. PPM1D p53-induced Ser/Thr protein phosphatase PPM1D (also known as ‘wild-type p53-inducible phosphatase 1’, Wip1) is a member of the PPM1 family. One of the functions of PPM1D is to negatively regulate the DNA damage response through the dephosphorylation and inactivation of p53, ATM, p38 and Chk1/2 [169–171]. PPM1D overexpression has been observed in many different types of human cancers (reviewed in Ref. [172]) and attenuates the effects of chemotherapy in patients with breast and ovarian carcinomas [173]. Thus it is likely that the over-expression of PPM1D participates in oncogenic transformation and cancer development. Furthermore, the decreased expression of PPM1D results in the enhancement of the DNA damage response [171]. In agreement with this, it has been shown that Ppm1d knockout mice show a tumor resistant phenotype. Specifically, Ppm1d knockout (Ppm1d −/−) mice are resistant to the development of spontaneous tumors over their entire lifespan [174], and show impaired mammary carcinogenesis relative to controls [170]. In addition, whereas transformation can be induced with a single oncogene in mouse embryonic fibroblasts (MEFs) from p53 knockout (Trp53 −/−) mice, injecting nude mice with Ppm1d −/−, Trp53 −/− MEFs expressing Hras1, Myc or Erbb2 did not result in tumor formation [170]. Following on from these findings, administration of Ppm1d inhibitors has shown to decrease the proliferation of xenograft tumors and tumors developed in MMTV/neu transgenic mice [175], suggesting that PPM1D is a potential target protein for cancer therapy. Indeed, much effort has gone into validating PPM1D as a therapeutic target and identifying specific inhibitors. 4.4.2. Poly(ADP-ribose) polymerase-1 Poly(ADP-ribose) polymerase-1 (PARP-1) and PARP-2 belong to a family of enzymes that use β-NAD + as a substrate to catalyze poly (ADP-ribosyl)ation of proteins. Their catalytic activity is stimulated by DNA-strand breaks (DSB) and thus they play a role in the DNA damage response, which has important consequences for genomic stability and tumor development (reviewed in Ref. [176]). Treatment of Parp-1 knockout (Parp-1 −/−) mice with alkylating agents reveals 324 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 extreme sensitivity and genomic instability as alkylating agents produce DNA damage repaired mainly through base excision repair (NER) and DSB repair pathways [177,178]. Conversely, Parp-1 −/− mice do not show elevated tumorigenesis to carcinogens which give rise to bulky DNA lesions [179] as they are repaired mainly through the NER pathway in which Parp-1 is not involved. Chemicallyinduced models of skin cancer have shown that development of DMBA/TPA-induced papilloma-like premalignant lesions is strongly delayed in Parp-1−/− mice with the final number of tumor-bearing mice and total tumor number being significantly reduced, relative to treated wildtype mice [180]. In agreement with this, pharmacologic inhibition of PARP-1 (using 3,4-dihydro-5-[4-(1-piperidinyl)butoxyl]1(2 H)-isoquinolinone) results in a strong delay in tumor formation and dramatic reduction in tumor size and multiplicity during DMBA/ TPA-induced skin carcinogenesis [181]. The authors proposed that PARP inhibition or genetic deletion of PARP-1 prevents tumor promotion through its ability to co-operate with the activation AP-1, NF-κB, and HIF-1α [181]. Interestingly, the same group found a gain in tumor-free survival in Parp-1 −/− mice on a p53-deficient background, which was confirmed by a significant delay in tumor formation in nude mice injected with Ras-transformed fibroblasts from Parp-1 −/−, p53−/− mice, and suggested that this was due to the abrogation of the oxydated status of p53 −/− cells (as iNOS expression and nitrite release were dramatically reduced in the Parp-1 −/−, p53 −/− mice) [182]. However, it needs to be mentioned that some studies have found an increased incidence of tumorigenesis in Parp-1 −/− mice in a DNA repair-deficient background. For example, in p53 heterozygous and homozygous backgrounds, Parp-1 −/− mice develop a variety of tumors, including mammary gland carcinoma, lung cancer, brain tumor and lymphoma, faster than Parp-1−/−, p53+/+ mice [183–185], and on a background of haploinsufficiency of non-homologous end-joining molecule Ku80 (Ku80+/− mice), a high frequency of liver cancer was observed in Parp-1 −/− mice relative to Parp-1 +/+ mice [186]. In addition, Parp-1 co-operated with Werner syndrome protein in the maintenance of chromosomal integrity and suppressing tumorigenesis [187]. Thus further studies are needed to examine the benefits of loss of PARP-1 in the context of a deficiency in certain types of DNA repair mechanisms and in specific tissues. Nevertheless it cannot be ignored that PARP-1 is over-expressed in many types of human tumors, frequently correlating with poor outcome (reviewed in Ref. [176]). Studies in mice have shown that genetic elimination of PARP-1 in malignant melanoma (RNAi-mediated depletion of Parp-1 in murine melanoma B16 cells) reduces tumor progression and chemoresistance in vivo [188], and PARP inhibition is particularly toxic in cancer cell lines and human tumors that lack BRCA1 or BRCA2 and are defective in homologous recombination [189,190]. Thus, PARP inhibitors that compete with β-NAD + have two therapeutic applications in cancer: (i) as chemo/radio-potentiators and (ii) as a stand-alone therapy for tumor types that are already deficient in certain types of DNA repair mechanisms (reviewed in Ref. [191]). 4.4.3. O 6-methylguanine-DNA methyltransferase Several environmental carcinogens and chemotherapeutic drugs alkylate DNA, and the most mutagenic lesion is the alkylation at the O 6-position of the DNA base guanine (O 6MeG). This class of mutagenic DNA adducts is repaired by the ‘suicide’ enzyme O 6methylguanine-DNA methyltransferase (MGMT), which transfers the alkyl group to a cysteine residue in its active site, after which the protein becomes inactive and targeted for proteasomal degradation. Thus MGMT is a key node in the defense against commonly found carcinogens, and a marker of resistance of normal and cancer cells exposed to alkylating therapeutics. MGMT also likely protects against therapy-related tumor formation caused by these highly mutagenic drugs (reviewed in Ref. [192]). The functional importance of MGMT in cancer prevention/resistance has been demonstrated by transgenic over-expression of human MGMT in mice. Numerous studies have shown that MGMT-CD2 transgenic mice, predominantly expressing MGMT not only in the thymus, were significantly more protected from developing tumors after being exposed to Nalkylated nitrosamines and their derivatives, specifically MNUinduced thymic lymphomas [193–196], AOM-induced colonic aberrant crypt foci [197], and NNK-induced lung tumors [198], compared with non-transgenic mice. Similarly, transgenic mice over-expressing MGMT under control of the cytokeratin promoter (Ck.MGMT mice) showed significantly lower numbers of malignant carcinomas than control mice after treatment of DMBA/TPA-induced benign skin papillomas with MNU [199]. Transgenic mice over-expressing an inducible form of the Escherichia coli MGMT gene, ada, showed a statistically significant reduction of liver tumor formation after treatment with hepatocarcinogen DEN [200], and a degree of protection against malignant progression of spontaneously developing liver tumors (as although there was no significant difference in tumor incidence compared to controls, ada-transgenic mice had fewer malignant tumors and survived longer) [201]. Further work from the same group has shown that the MGMT transgene partially rescues the cancer-prone phenotype of MNU-treated mice either deficient in the DNA mismatch repair gene Pms2 [202], heterozygous for p53 [203], or over-expressing LMO1 [204]. In addition, the spontaneous development of hepatocellular carcinoma in C3HeB/FeJ male mice was significantly reduced in those expressing human MGMT compared with non-transgenics [205]. 4.4.4. DNA methyltransferase 1 DNA methylation is the reversible addition of a methyl group to cytosine in CpG dinucleotides and is essential for normal embryonic development. However, global genomic hypomethylation and aberrant hypermethylation of regulatory regions of tumor suppressor genes have been associated with chromosomal instability and transcription repression, respectively, providing cancer cells with a selective advantage (reviewed in Ref. [206]). DNA methyltransferases are the enzymes responsible for the addition of methyl groups to CpG dinucleotides, which, together with histone modifiers, initiate the events necessary for transcription repression to occur. The methylation of genomic DNA in cancer cells is catalyzed by DNA methyltransferases DNMT1 and DNMT3B, revealing significantly elevated expression in different types of cancers. In the case of colo-rectal cancer, mouse models have demonstrated a strong link between hypthermethylation-induced silencing of tumor suppressor genes and tumorigenesis. For example, mice heterozygous for a targeted Dnmt1 allele (Dnmt1 +/−) showed a retarded net growth rate of intestinal adenomas and reduced tumor multiplicity by approximately 50% in ApcMin/+ mice [207], and Dnmt1+/−, Apc+/Min mice given a weekly dose of the DNA methyltransferase inhibitor 5-aza-deoxycytidine, show reduced average numbers of intestinal adenomas [208]. Mouse models carrying hypomorphic alleles of Dnmt1 showed a variety of tumor resistance phenotypes, including complete suppression of polyp formation in ApcMin/+ mice [209], reduced intestinal tumor formation in Mlh1−/− mice [210] and reduced tumor burden (acinar cell pancreatic cancer) in ApcMin/+, p53−/− mice [211]. However, it must be noted that Dnmt1−/−, Mlh1−/− mice developed invasive T- and B-cell lymphomas earlier and at a much higher frequency than Dnmt+/+, Mlh1−/− littermates [210]. Thus, the reduction of Dnmt1 activity has significant but opposing outcomes in the development of two different tumor types: DNA hypomethylation and mismatch repair deficiency interact to exacerbate lymphomagenesis, while hypomethylation protects against intestinal tumors. 4.4.5. Terc Telomerase is a ribonucleoprotein enzyme complex that adds telomeric repeats to the ends of chromosomes. The core telomerase L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 components are the telomerase reverse transcriptase (TERT) catalytic subunit, encoded by the TERT gene on chromosome 5p15, and the telomerase RNA (TR) template subunit, encoded by the TERC gene on chromosome 3q26. Telomerase is frequently over-expressed in many cancers, a consequence of which is maintenance of telomere length, and thus evasion of the steady decrease in telomere length that accompanies proliferation of normal cells, although it has also been suggested that telomerase up-regulation confers other advantages on cancer cells independent of its enzymatic activity (reviewed in Ref. [212]). Late-generation (G5) Terc −/− mice, which have short telomeres and are telomerase-deficient, show defects in proliferative tissues and a moderate increase in the incidence of spontaneous tumors in highly proliferative cell types (lymphomas, teratocarcinomas), presumably as a consequence of chromosomal instability [213]. However, different cell types vary in their sensitivity to the chromosomal instability produced by telomere loss or to the activation of telomere-rescue mechanisms as evidenced by the fact that G5 Terc −/− mice are resistant to tumor development in chemicalinduced carcinogenesis in the skin (DMBA/TPA-induced skin papillomas regressed within one week after termination of TPA treatment) [213] and liver (significantly decreased number and size of CCl4 or DEN-induced surface liver nodules) [214]. Similarly, Terc −/− mice were found to be relatively resistant to spontaneous tumorigenesis in cancer-prone mice, including Ink4a −/− mice [215], Apc Min/+ mice [216], Alb-uPA transgenic mice [214], Atm −/− mice [217], p53 R172P/ R172P mice [218], and MMT transgenic mice (expressing the MUC1 (Mucin 1) and polyomavirus middle T (PyMT) oncogene) [219]. In addition, transplant and xenograft studies have demonstrated that reduction of Terc expression, via the use of double-stranded oligodeoxynucleotides, ribozymes, or antisense vectors, can reduce the in vivo tumorigenicity of liver and gastric cancer cell lines [220,221] and metastatic potential of melanoma cell lines [222,223]. However, it needs to be noted that G5 Terc −/− mice show defects in proliferative tissues and a moderate increase in the incidence of spontaneous tumors in highly proliferative cell types (lymphomas, teratocarcinomas), presumably as a consequence of chromosomal instability in these mice [213]. Other studies have found that G5 Terc −/− mice also show an orally-dosed DMBA-induced metastatic tumor burden similar to that of wildtype mice [224]. 4.5. Miscellaneous 4.5.1. Eph receptor A2 The Eph receptor tyrosine kinase (RTK) family is composed of 14 members that are subdivided into two subclasses based upon their complementary Ephrin ligands (there are nine Ephrin ligands classified as either Ephrin A or Ephrin B). The Eph RTK and their cell-surface bound ligands are involved in a variety of cell-to-cell inter-communications that affect processes such as cell patterning, guidance, migration and adhesion. High expression levels of one member of the Eph receptors, EphA2 on human chromosome 1p36.1, are correlated with tumor metastasis and poor prognosis [225]. Confirming the role of EphA2 in malignant invasion, human gastric cancer SGC-7901 cell xenografts in nude mice given intratumoral injections of EphA2 siRNA plasmid showed suppressed growth (the tumors also showed reduced expression of matrix metalloproteinase-9) [226]. Expression of EphA2 mutants (that lack the cytoplasmic domain or carry a point mutation that inhibits its kinase activity) in breast cancer cells resulted in decreased tumor volume, increased tumor apoptosis in primary tumors, and significantly reduced lung metastases in both experimental and spontaneous metastasis models, suggesting that receptor phosphorylation and kinase activity of the EphA2 receptor, at least in part, contribute to tumor malignancy [227]. In addition, there is data to suggest that EphA2 co-operates with ErbB2 to promote tumor progression in mice, and importantly that EphA2 function in tumor progression depends on oncogene context, as tumors from 325 MMTV/neu (which overexpress neu, the rat homolog of ErbB2, in the mammary epithelium) but not MMTV/PyV-mT mice (which overexpress the polyomavirus middle TAg in mammary epithelium) were sensitive to therapeutic inhibition of EphA2 [228]. Mouse models of GI cancer have demonstrated that EphA2 plays an oncogenic role in the intestine as ApcMin/+ mice carrying a knockout allele of EphA2 gene develop significantly fewer intestinal tumors [229], yet there is evidence that EphA2 may function as a tumor suppressor, as mice that are homozygous for a gene trap allele of EphA2 (EphA2−/−) demonstrate increased susceptibility to skin carcinogenesis [230]. Thus whether EphA2 acts as an oncogene or tumor suppressor may depend on a multitude of complex factors, such as the tissue type, normal versus malignant cells, presence/absence of ligands or endogenous receptor tyrosine kinases, and co-operation with other specific oncogenic mutations. 4.5.2. Fibroblast factor binding protein The fibroblast growth factor (FGF) family consists of at least 23 structurally related polypeptide growth factors that are important regulators of angiogenesis, embryonic development, cell migration, proliferation and differentiation, and are particularly important in tumor growth. One mechanism that determines their biological activities in tumor growth is based on their mobilization from the extracellular matrix, which can occur via heparanase-mediated cleavage of heparansulphate proteoglycans or the action of the fibroblast growth factor-binding protein (FGF-BP) serving as a carrier protein. FGF-BP expression is highly tissue specific and strictly regulated through different promoter elements. Besides its role in embryogenesis and wound healing, FGF-BP is upregulated in several tumors and it is associated especially with early stages of tumor formation, where angiogenesis plays a critical role (reviewed in Ref. [231]). In agreement with this, several mouse tumor models have shown that targeting of FGF-BP abolishes or reduces tumor growth and angiogenesis. For example, human squamous cell carcinoma ME-180 and colon cancer LS174T cell lines depleted of FGF-BP by targeting with specific ribozymes exhibited reduced growth and angiogenesis when administered to nude mice [232]. Similarly, administration of FGF-BP-specific siRNA duplexes to tumor xenograft-bearing mice lead to an ~ 40% reduced tumor growth relative to controls [233]. In addition, it was found that adenoviral vector‐mediated wildtype p53 transduction resulted in growth inhibition of squamous cell carcinoma of the head and neck tumor cells in a xenograft mouse model, that was due, at least in part, to anti-angiogenesis mediated by the down-modulation of FGF-BP [234]. Finally, in vivo all-trans retinoic acid treatment of squamous cell carcinoma xenografts in nude mice reduced FGF-BP expression in parallel with decreased tumor growth and angiogenesis and induction of apoptosis [235]. This indicates that FGF-BP can be rate-limiting for tumor growth and serves as an angiogenic switch molecule. 5. Conclusions The mouse has proved itself to be a most invaluable tool to researchers in understanding the genetics of cancer susceptibility and resistance. The availability of different strains of wildtype mice, spontaneously occurring mutants and GM mice have shown that there are many genes involved in the cancer resistance phenotype. In general, genes that augment the host immune system; and/or dampen down the inflammatory response; and/or halt proliferation and promote apoptosis of the tumor cells; and/or maintain the integrity of the genome all play important roles in cancer resistance. However, investigation of these genes using mouse models has revealed a subtlety to the situation with some genes being involved in both tumor resistance and susceptibility, depending on their context, such as tissue type, relative concentration of the protein or genetic background of the mouse. In addition, some genes only confer resistance to specific types of tumors, such as carcinogen-induced tumors but not oncogene- 326 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 induced tumors. Thus the mouse has played a crucial role in highlighting the complexities of identifying genes responsible for cancer resistance, as well as identifying potential targets for chemoprevention. Conflict of interest The authors declare no conflict of interest. Acknowledgements D.J.A was supported by Cancer Research UK and the Wellcome Trust. References [1] G.B. Challis, H.J. Stam, The spontaneous regression of cancer. A review of cases from 1900 to 1987, Acta Oncol. 29 (1990) (1900) 545–550. [2] B.A. Ponder, Inherited predisposition to cancer, Trends Genet. 6 (1990) 213–218. [3] A. Balmain, H. Nagase, Cancer resistance genes in mice: models for the study of tumour modifiers, Trends Genet. 14 (1998) 139–144. [4] M.B. Shimkin, G.D. Stoner, Lung tumors in mice: application to carcinogenesis bioassay, Adv. Cancer Res. 21 (1975) 1–58. [5] D. Sun, H. Ren, M. Oertel, R.S. Sellers, D.A. Shafritz, L. Zhu, Inactivation of p27Kip1 promotes chemical mouse liver tumorigenesis in the resistant strain C57BL/6J, Mol. Carcinog. 47 (2008) 47–55. [6] A. Papanikolaou, Q.S. Wang, D.A. Delker, D.W. Rosenberg, Azoxymethaneinduced colon tumors and aberrant crypt foci in mice of different genetic susceptibility, Cancer Lett. 130 (1998) 29–34. [7] F. Lilly, T. Pincus, Genetic control of murine viral leukemogenesis, Adv. Cancer Res. 17 (1973). [8] F. Lilly, The role of genetics in Gross virus leukemogenesis, Bibl. Haematol. (1970) 213–220. [9] F. Lilly, E.A. Boyse, L.J. Old, Genetic basis of susceptibility to viral leukaemogenesis, Lancet 2 (1964) 1207–1209. [10] J.R. Tennant, G.D. Snell, The H-2 locus and viral leukemogenesis as studied in congenic strains of mice, J. Natl. Cancer Inst. 41 (1968) 597–604. [11] S. Nandi, The histocompatability-2 locus and susceptibility to bittner virus borne by red blood cells in mice, Proc. Natl. Acad. Sci. U. S. A. 58 (1967) 485–492. [12] B. Chesebro, K. Wehrly, J. Stimpfling, Host genetic control of recovery from Friend leukemia virus-induced splenomegaly: mapping of a gene within the major histocompatability complex, J. Exp. Med. 140 (1974) 1457–1467. [13] A. Goldfeder, Urethan and x-ray effects on mice of a tumor-resistant strain, X-Gf, Cancer Res. 32 (1972) 2771–2777. [14] J.L. Guenet, F. Bonhomme, Wild mice: an ever-increasing contribution to a popular mammalian model, Trends Genet. 19 (2003) 24–31. [15] R. Bremner, A. Balmain, Genetic changes in skin tumor progression: correlation between presence of a mutant ras gene and loss of heterozygosity on mouse chromosome 7, Cell 61 (1990) 407–417. [16] G.H. Lee, L.M. Bennett, R.A. Carabeo, N.R. Drinkwater, Identification of hepatocarcinogen-resistance genes in DBA/2 mice, Genetics 139 (1995) 387–395. [17] T. van Wezel, A.P. Stassen, C.J. Moen, A.A. Hart, M.A. van der Valk, P. Demant, Gene interaction and single gene effects in colon tumour susceptibility in mice, Nat. Genet. 14 (1996) 468–470. [18] R.J. Fijneman, S.S. de Vries, R.C. Jansen, P. Demant, Complex interactions of new quantitative trait loci, Sluc1, Sluc2, Sluc3, and Sluc4, that influence the susceptibility to lung cancer in the mouse, Nat. Genet. 14 (1996) 465–467. [19] R. Buffenstein, The naked mole-rat: a new long-living model for human aging research, J. Gerontol. A Biol. Sci. Med. Sci. 60 (2005) 1369–1377. [20] A. Seluanov, C. Hine, J. Azpurua, M. Feigenson, M. Bozzella, Z. Mao, K.C. Catania, V. Gorbunova, Hypersensitivity to contact inhibition provides a clue to cancer resistance of naked mole-rat, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 19352–19357. [21] S. Liang, J. Mele, Y. Wu, R. Buffenstein, P.J. Hornsby, Resistance to experimental tumorigenesis in cells of a long-lived mammal, the naked mole-rat (Heterocephalus glaber), Aging Cell 9 (2010) 626–635. [22] I.K. Egorov, A mutation of the histocompatibility-2 locus in the mouse, Genetika 3 (1967) 136–144. [23] D.W. Bailey, H.I. Kohn, Inherited histocompatibility changes in progeny of irradiated and unirradiated inbred mice, Genet. Res. 6 (1965) 330–340. [24] Y.H. Sun, R.S. Goodenow, L. Hood, Molecular basis of the dm1 mutation in the major histocompatibility complex of the mouse: a D/L hybrid gene, J. Exp. Med. 162 (1985) 1588–1602. [25] M. Miyazawa, J. Nishio, B. Chesebro, Protection against Friend retrovirusinduced leukemia by recombinant vaccinia viruses expressing the gag gene, J. Virol. 66 (1992) 4497–4507. [26] I.K. Egorov, O.S. Egorov, Detection of new MHC mutations in mice by skin grafting, tumor transplantation and monoclonal antibodies: a comparison, Genetics 118 (1988) 287–298. [27] O.S. Egorov, Y. Liu, E.E. Sargent, G.I. Drizlikh, I.K. Egorov, Screening mouse mutations for resistance to cancer metastasis, Immunogenetics 36 (1992) 219–223. [28] I.K. Egorov, B.V. Nickonenko, Genes for control of spread of the tumor and mycobacterium tuberculosis infection in the mouse, Crit. Rev. Immunol. 14 (1994) 337–353. [29] Y. Liu, D.L. McMinimy, A.Y. Savinov, K.A. Johnson, S.G. Kremlev, A.I. Chapoval, I.K. Egorov, Hidden variables: unstable Abeta chain genes encoding antigen recognition structures in tumor survivors, Mol. Immunol. 37 (2000) 1091–1110. [30] Z. Cui, M.C. Willingham, A.M. Hicks, M.A. Alexander-Miller, T.D. Howard, G.A. Hawkins, M.S. Miller, H.M. Weir, W. Du, C.J. DeLong, Spontaneous regression of advanced cancer: identification of a unique genetically determined, agedependent trait in mice, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 6682–6687. [31] J. Koch, A. Boschian, J. Hau, K. Rieneck, Frequency of the cancer-resistant phenotype in SR/CR mice and the effect of litter seriation, In Vivo 22 (2008) 565–569. [32] G. Riedlinger, J. Adams, J.R. Stehle Jr., M.J. Blanks, A.M. Sanders, A.M. Hicks, M.C. Willingham, Z. Cui, The spectrum of resistance in SR/CR mice: the critical role of chemoattraction in the cancer/leukocyte interaction, BMC Cancer 10 (2010) 179. [33] A.M. Hicks, M.C. Willingham, W. Du, C.S. Pang, L.J. Old, Z. Cui, Effector mechanisms of the anti-cancer immune responses of macrophages in SR/CR mice, Cancer Immun. 6 (2006) 11. [34] A.M. Hicks, G. Riedlinger, M.C. Willingham, M.A. Alexander-Miller, C. Von KapHerr, M.J. Pettenati, A.M. Sanders, H.M. Weir, W. Du, J. Kim, A.J. Simpson, L.J. Old, Z. Cui, Transferable anticancer innate immunity in spontaneous regression/complete resistance mice, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 7753–7758. [35] P. Klatt, M. Serrano, Engineering cancer resistance in mice, Carcinogenesis 24 (2003) 817–826. [36] A.K. Abbas, K.M. Murphy, A. Sher, Functional diversity of helper T lymphocytes, Nature 383 (1996) 787–793. [37] F. Pages, J. Galon, M.C. Dieu-Nosjean, E. Tartour, C. Sautes-Fridman, W.H. Fridman, Immune infiltration in human tumors: a prognostic factor that should not be ignored, Oncogene 29 (2010) 1093–1102. [38] M. Chiyo, O. Shimozato, L. Yu, K. Kawamura, T. Iizasa, T. Fujisawa, M. Tagawa, Expression of IL-27 in murine carcinoma cells produces antitumor effects and induces protective immunity in inoculated host animals, Int. J. Cancer 115 (2005) 437–442. [39] S. Ugai, O. Shimozato, L. Yu, Y.Q. Wang, K. Kawamura, H. Yamamoto, T. Yamaguchi, H. Saisho, S. Sakiyama, M. Tagawa, Transduction of the IL-21 and IL-23 genes in human pancreatic carcinoma cells produces natural killer celldependent and -independent antitumor effects, Cancer Gene Ther. 10 (2003) 771–778. [40] M.J. Brunda, L. Luistro, R.R. Warrier, R.B. Wright, B.R. Hubbard, M. Murphy, S.F. Wolf, M.K. Gately, Antitumor and antimetastatic activity of interleukin 12 against murine tumors, J. Exp. Med. 178 (1993) 1223–1230. [41] J.P. Zou, N. Yamamoto, T. Fujii, H. Takenaka, M. Kobayashi, S.H. Herrmann, S.F. Wolf, H. Fujiwara, T. Hamaoka, Systemic administration of rIL-12 induces complete tumor regression and protective immunity: response is correlated with a striking reversal of suppressed IFN-gamma production by anti-tumor T cells, Int. Immunol. 7 (1995) 1135–1145. [42] M.J. Robertson, J. Ritz, Interleukin 12: basic biology and potential applications in cancer treatment, Oncologist 1 (1996) 88–97. [43] S.M. Meeran, S.K. Mantena, S. Meleth, C.A. Elmets, S.K. Katiyar, Interleukin-12deficient mice are at greater risk of UV radiation-induced skin tumors and malignant transformation of papillomas to carcinomas, Mol. Cancer Ther. 5 (2006) 825–832. [44] S.D. Sharma, S.M. Meeran, N. Katiyar, G.B. Tisdale, N. Yusuf, H. Xu, C.A. Elmets, S.K. Katiyar, IL-12 deficiency suppresses 12-O-tetradecanoylphorbol-13-acetateinduced skin tumor development in 7,12-dimethylbenz(a)anthracene-initiated mouse skin through inhibition of inflammation, Carcinogenesis 30 (2009) 1970–1977. [45] E.A. Carswell, L.J. Old, R.L. Kassel, S. Green, N. Fiore, B. Williamson, An endotoxininduced serum factor that causes necrosis of tumors, Proc. Natl. Acad. Sci. U. S. A. 72 (1975) 3666–3670. [46] F. Guadagni, P. Ferroni, R. Palmirotta, I. Portarena, V. Formica, M. Roselli, Review. TNF/VEGF cross-talk in chronic inflammation-related cancer initiation and progression: an early target in anticancer therapeutic strategy, In Vivo 21 (2007) 147–161. [47] M. Suganuma, S. Okabe, M.W. Marino, A. Sakai, E. Sueoka, H. Fujiki, Essential role of tumor necrosis factor alpha (TNF-alpha) in tumor promotion as revealed by TNF-alpha-deficient mice, Cancer Res. 59 (1999) 4516–4518. [48] R.J. Moore, D.M. Owens, G. Stamp, C. Arnott, F. Burke, N. East, H. Holdsworth, L. Turner, B. Rollins, M. Pasparakis, G. Kollias, F. Balkwill, Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis, Nat. Med. 5 (1999) 828–831. [49] K.A. Scott, R.J. Moore, C.H. Arnott, N. East, R.G. Thompson, B.J. Scallon, D.J. Shealy, F.R. Balkwill, An anti-tumor necrosis factor-alpha antibody inhibits the development of experimental skin tumors, Mol. Cancer Ther. 2 (2003) 445–451. [50] F. Balkwill, Tumor necrosis factor or tumor promoting factor? Cytokine Growth Factor Rev. 13 (2002) 135–141. [51] B. Knight, G.C. Yeoh, K.L. Husk, T. Ly, L.J. Abraham, C. Yu, J.A. Rhim, N. Fausto, Impaired preneoplastic changes and liver tumor formation in tumor necrosis factor receptor type 1 knockout mice, J. Exp. Med. 192 (2000) 1809–1818. [52] F. Chang, M.R. Lacey, M. Bouljihad, K.H. Bentrup, I.S. Fortgang, Tumor necrosis factor receptor 1 functions as a tumor suppressor, Am. J. Physiol. Gastrointest. Liver Physiol. 302 (2012) G195–G206. L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 [53] M.L. Alegre, K.A. Frauwirth, C.B. Thompson, T-cell regulation by CD28 and CTLA4, Nat. Rev. Immunol. 1 (2001) 220–228. [54] V.A. Boussiotis, J.G. Gribben, G.J. Freeman, L.M. Nadler, Blockade of the CD28 costimulatory pathway: a means to induce tolerance, Curr. Opin. Immunol. 6 (1994) 797–807. [55] S. Beissert, J.A. Bluestone, I. Mindt, M. Voskort, D. Metze, A. Mehling, T.A. Luger, T. Schwarz, S. Grabbe, Reduced ultraviolet-induced carcinogenesis in mice with a functional disruption in B7-mediated costimulation, J. Immunol. 163 (1999) 6725–6731. [56] V.K. Kuchroo, M.P. Das, J.A. Brown, A.M. Ranger, S.S. Zamvil, R.A. Sobel, H.L. Weiner, N. Nabavi, L.H. Glimcher, B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy, Cell 80 (1995) 707–718. [57] C.D. Funk, Prostaglandins and leukotrienes: advances in eicosanoid biology, Science 294 (2001) 1871–1875. [58] J.E. Burke, E.A. Dennis, Phospholipase A2 structure/function, mechanism, and signaling, J. Lipid Res. 50 (Suppl.) (2009) S237–S242. [59] R.J. Fijneman, R.T. Cormier, The roles of sPLA2-IIA (Pla2g2a) in cancer of the small and large intestine, Front. Biosci. 13 (2008) 4144–4174. [60] L.K. Su, K.W. Kinzler, B. Vogelstein, A.C. Preisinger, A.R. Moser, C. Luongo, K.A. Gould, W.F. Dove, Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene, Science 256 (1992) 668–670. [61] W.F. Dietrich, E.S. Lander, J.S. Smith, A.R. Moser, K.A. Gould, C. Luongo, N. Borenstein, W. Dove, Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse, Cell 75 (1993) 631–639. [62] R.T. Cormier, A. Bilger, A.J. Lillich, R.B. Halberg, K.H. Hong, K.A. Gould, N. Borenstein, E.S. Lander, W.F. Dove, The Mom1AKR intestinal tumor resistance region consists of Pla2g2a and a locus distal to D4Mit64, Oncogene 19 (2000) 3182–3192. [63] R.T. Cormier, K.H. Hong, R.B. Halberg, T.L. Hawkins, P. Richardson, R. Mulherkar, W.F. Dove, E.S. Lander, Secretory phospholipase Pla2g2a confers resistance to intestinal tumorigenesis, Nat. Genet. 17 (1997) 88–91. [64] K.A. Silverman, R. Koratkar, L.D. Siracusa, A.M. Buchberg, Identification of the modifier of Min 2 (Mom2) locus, a new mutation that influences Apc-induced intestinal neoplasia, Genome Res. 12 (2002) 88–97. [65] A.A. Baran, K.A. Silverman, J. Zeskand, R. Koratkar, A. Palmer, K. McCullen, W.J. Curran Jr., T.B. Edmonston, L.D. Siracusa, A.M. Buchberg, The modifier of Min 2 (Mom2) locus: embryonic lethality of a mutation in the Atp5a1 gene suggests a novel mechanism of polyp suppression, Genome Res. 17 (2007) 566–576. [66] L.N. Kwong, A. Shedlovsky, B.S. Biehl, L. Clipson, C.A. Pasch, W.F. Dove, Identification of Mom7, a novel modifier of Apc(Min/+) on mouse chromosome 18, Genetics 176 (2007) 1237–1244. [67] R.J. Fijneman, J.R. Peham, M.A. van de Wiel, G.A. Meijer, I. Matise, A. Velcich, R.T. Cormier, Expression of Pla2g2a prevents carcinogenesis in Muc2-deficient mice, Cancer Sci. 99 (2008) 2113–2119. [68] R.J. Fijneman, L.K. Bade, J.R. Peham, M.A. van de Wiel, V.W. van Hinsbergh, G.A. Meijer, M.G. O'Sullivan, R.T. Cormier, Pla2g2a attenuates colon tumorigenesis in azoxymethane-treated C57BL/6 mice; expression studies reveal Pla2g2a target genes and pathways, Cell. Oncol. 31 (2009) 345–356. [69] K.H. Hong, J.C. Bonventre, E. O'Leary, J.V. Bonventre, E.S. Lander, Deletion of cytosolic phospholipase A(2) suppresses Apc(Min)-induced tumorigenesis, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 3935–3939. [70] K. Takaku, M. Sonoshita, N. Sasaki, N. Uozumi, Y. Doi, T. Shimizu, M.M. Taketo, Suppression of intestinal polyposis in Apc(delta 716) knockout mice by an additional mutation in the cytosolic phospholipase A(2) gene, J. Biol. Chem. 275 (2000) 34013–34016. [71] E.M. Smyth, T. Grosser, M. Wang, Y. Yu, G.A. FitzGerald, Prostanoids in health and disease, J. Lipid Res. 50 (Suppl.) (2009) S423–S428. [72] D. Wang, R.N. Dubois, Eicosanoids and cancer, nature reviews, Cancer 10 (2010) 181–193. [73] F.V. Din, E. Theodoratou, S.M. Farrington, A. Tenesa, R.A. Barnetson, R. Cetnarskyj, L. Stark, M.E. Porteous, H. Campbell, M.G. Dunlop, Effect of aspirin and NSAIDs on risk and survival from colorectal cancer, Gut 59 (2010) 1670–1679. [74] M. Oshima, J.E. Dinchuk, S.L. Kargman, H. Oshima, B. Hancock, E. Kwong, J.M. Trzaskos, J.F. Evans, M.M. Taketo, Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2), Cell 87 (1996) 803–809. [75] P.C. Chulada, M.B. Thompson, J.F. Mahler, C.M. Doyle, B.W. Gaul, C. Lee, H.F. Tiano, S.G. Morham, O. Smithies, R. Langenbach, Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces intestinal tumorigenesis in Min mice, Cancer Res. 60 (2000) 4705–4708. [76] X. Zhang, S.G. Morham, R. Langenbach, R.B. Baggs, D.A. Young, Lack of cyclooxygenase-2 inhibits growth of teratocarcinomas in mice, Exp. Cell Res. 254 (2000) 232–240. [77] H.F. Tiano, C.D. Loftin, J. Akunda, C.A. Lee, J. Spalding, A. Sessoms, D.B. Dunson, E.G. Rogan, S.G. Morham, R.C. Smart, R. Langenbach, Deficiency of either cyclooxygenase (COX)-1 or COX-2 alters epidermal differentiation and reduces mouse skin tumorigenesis, Cancer Res. 62 (2002) 3395–3401. [78] N.L. Barnes, F. Warnberg, G. Farnie, D. White, W. Jiang, E. Anderson, N.J. Bundred, Cyclooxygenase-2 inhibition: effects on tumour growth, cell cycling and lymphangiogenesis in a xenograft model of breast cancer, Br. J. Cancer 96 (2007) 575–582. [79] K. Muller-Decker, G. Neufang, I. Berger, M. Neumann, F. Marks, G. Furstenberger, Transgenic cyclooxygenase-2 overexpression sensitizes mouse skin for carcinogenesis, Proc. Natl. Acad. Sci. U. S. A. 99 (2002) 12483–12488. 327 [80] D.K. Bol, R.B. Rowley, C.P. Ho, B. Pilz, J. Dell, M. Swerdel, K. Kiguchi, S. Muga, R. Klein, S.M. Fischer, Cyclooxygenase-2 overexpression in the skin of transgenic mice results in suppression of tumor development, Cancer Res. 62 (2002) 2516–2521. [81] J.E. Rundhaug, A. Pavone, E. Kim, S.M. Fischer, The effect of cyclooxygenase-2 overexpression on skin carcinogenesis is context dependent, Mol. Carcinog. 46 (2007) 981–992. [82] K. Sonzogni-Desautels, D.W. Knapp, E. Sartin, M. Dore, Effect of cyclooxygenase inhibitors in a xenograft model of canine mammary tumours, Vet. Comp. Oncol. 9 (2011) 161–171. [83] J.R. Kanwar, R.K. Kanwar, H. Burrow, S. Baratchi, Recent advances on the roles of NO in cancer and chronic inflammatory disorders, Curr. Med. Chem. 16 (2009) 2373–2394. [84] R.G. Knowles, S. Moncada, Nitric oxide synthases in mammals, Biochem. J. 298 (Pt 2) (1994) 249–258. [85] S. Singh, A.K. Gupta, Nitric oxide: role in tumour biology and iNOS/NO-based anticancer therapies, Cancer Chemother. Pharmacol. 67 (2011) 1211–1224. [86] B. Ahn, H. Ohshima, Suppression of intestinal polyposis in Apc(Min/+) mice by inhibiting nitric oxide production, Cancer Res. 61 (2001) 8357–8360. [87] H.F. Yerushalmi, D.G. Besselsen, N.A. Ignatenko, K.A. Blohm-Mangone, J.L. Padilla-Torres, D.E. Stringer, H. Cui, H. Holubec, C.M. Payne, E.W. Gerner, The role of NO synthases in arginine-dependent small intestinal and colonic carcinogenesis, Mol. Carcinog. 45 (2006) 93–105. [88] D.J. Scott, M.A. Hull, E.J. Cartwright, W.K. Lam, A. Tisbury, R. Poulsom, A.F. Markham, C. Bonifer, P.L. Coletta, Lack of inducible nitric oxide synthase promotes intestinal tumorigenesis in the Apc(Min/+) mouse, Gastroenterology 121 (2001) 889–899. [89] D.N. Seril, J. Liao, G.Y. Yang, Colorectal carcinoma development in inducible nitric oxide synthase-deficient mice with dextran sulfate sodium-induced ulcerative colitis, Mol. Carcinog. 46 (2007) 341–353. [90] A. Chariot, The NF-kappaB-independent functions of IKK subunits in immunity and cancer, Trends Cell Biol. 19 (2009) 404–413. [91] F.R. Greten, L. Eckmann, T.F. Greten, J.M. Park, Z.W. Li, L.J. Egan, M.F. Kagnoff, M. Karin, IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer, Cell 118 (2004) 285–296. [92] E. Park, F. Zhu, B. Liu, X. Xia, J. Shen, T. Bustos, S.M. Fischer, Y. Hu, Reduction in IkappaB kinase alpha expression promotes the development of skin papillomas and carcinomas, Cancer Res. 67 (2007) 9158–9168. [93] B. Liu, E. Park, F. Zhu, T. Bustos, J. Liu, J. Shen, S.M. Fischer, Y. Hu, A critical role for I kappaB kinase alpha in the development of human and mouse squamous cell carcinomas, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 17202–17207. [94] J. Whelan, M.F. McEntee, Dietary (n-6) PUFA and intestinal tumorigenesis, J. Nutr. 134 (2004) 3421S–3426S. [95] J.X. Kang, J. Wang, L. Wu, Z.B. Kang, Transgenic mice: fat-1 mice convert n-6 to n-3 fatty acids, Nature 427 (2004) 504. [96] C.A. Hudert, K.H. Weylandt, Y. Lu, J. Wang, S. Hong, A. Dignass, C.N. Serhan, J.X. Kang, Transgenic mice rich in endogenous omega-3 fatty acids are protected from colitis, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 11276–11281. [97] J. Nowak, K.H. Weylandt, P. Habbel, J. Wang, A. Dignass, J.N. Glickman, J.X. Kang, Colitis-associated colon tumorigenesis is suppressed in transgenic mice rich in endogenous n-3 fatty acids, Carcinogenesis 28 (2007) 1991–1995. [98] K.H. Weylandt, L.F. Krause, B. Gomolka, C.Y. Chiu, S. Bilal, A. Nadolny, S.F. Waechter, A. Fischer, M. Rothe, J.X. Kang, Suppressed liver tumorigenesis in fat-1 mice with elevated omega-3 fatty acids is associated with increased omega-3 derived lipid mediators and reduced TNF-alpha, Carcinogenesis 32 (2011) 897–903. [99] S. Xia, Y. Lu, J. Wang, C. He, S. Hong, C.N. Serhan, J.X. Kang, Melanoma growth is reduced in fat-1 transgenic mice: impact of omega-6/omega-3 essential fatty acids, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 12499–12504. [100] A.J. Levine, p53, the cellular gatekeeper for growth and division, Cell 88 (1997) 323–331. [101] T. Soussi, G. Lozano, p53 mutation heterogeneity in cancer, Biochem. Biophys. Res. Commun. 331 (2005) 834–842. [102] L.A. Donehower, Does p53 affect organismal aging? J. Cell. Physiol. 192 (2002) 23–33. [103] L.A. Donehower, G. Lozano, 20 years studying p53 functions in genetically engineered mice, nature reviews, Cancer 9 (2009) 831–841. [104] S.D. Tyner, S. Venkatachalam, J. Choi, S. Jones, N. Ghebranious, H. Igelmann, X. Lu, G. Soron, B. Cooper, C. Brayton, S. Hee Park, T. Thompson, G. Karsenty, A. Bradley, L.A. Donehower, p53 mutant mice that display early ageingassociated phenotypes, Nature 415 (2002) 45–53. [105] B. Maier, W. Gluba, B. Bernier, T. Turner, K. Mohammad, T. Guise, A. Sutherland, M. Thorner, H. Scrable, Modulation of mammalian life span by the short isoform of p53, Genes Dev. 18 (2004) 306–319. [106] I. Garcia-Cao, M. Garcia-Cao, J. Martin-Caballero, L.M. Criado, P. Klatt, J.M. Flores, J.C. Weill, M.A. Blasco, M. Serrano, “Super p53” mice exhibit enhanced DNA damage response, are tumor resistant and age normally, EMBO J. 21 (2002) 6225–6235. [107] D.A. Greenhalgh, X.J. Wang, L.A. Donehower, D.R. Roop, Paradoxical tumor inhibitory effect of p53 loss in transgenic mice expressing epidermal-targeted vrasHa, v-fos, or human transforming growth factor alpha, Cancer Res. 56 (1996) 4413–4423. [108] C.J. Sherr, Divorcing ARF and p53: an unsettled case, nature reviews, Cancer 6 (2006) 663–673. [109] W.Y. Kim, N.E. Sharpless, The regulation of INK4/ARF in cancer and aging, Cell 127 (2006) 265–275. 328 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 [110] A. Matheu, C. Pantoja, A. Efeyan, L.M. Criado, J. Martin-Caballero, J.M. Flores, P. Klatt, M. Serrano, Increased gene dosage of Ink4a/Arf results in cancer resistance and normal aging, Genes Dev. 18 (2004) 2736–2746. [111] A. Matheu, A. Maraver, P. Klatt, I. Flores, I. Garcia-Cao, C. Borras, J.M. Flores, J. Vina, M.A. Blasco, M. Serrano, Delayed ageing through damage protection by the Arf/p53 pathway, Nature 448 (2007) 375–379. [112] L.P. Weng, J.L. Brown, C. Eng, PTEN coordinates G(1) arrest by down-regulating cyclin D1 via its protein phosphatase activity and up-regulating p27 via its lipid phosphatase activity in a breast cancer model, Hum. Mol. Genet. 10 (2001) 599–604. [113] A. Suzuki, J.L. de la Pompa, V. Stambolic, A.J. Elia, T. Sasaki, I. del Barco Barrantes, A. Ho, A. Wakeham, A. Itie, W. Khoo, M. Fukumoto, T.W. Mak, High cancer susceptibility and embryonic lethality associated with mutation of the PTEN tumor suppressor gene in mice, Curr. Biol. 8 (1998) 1169–1178. [114] J. Dupont, J.P. Renou, M. Shani, L. Hennighausen, D. LeRoith, PTEN overexpression suppresses proliferation and differentiation and enhances apoptosis of the mouse mammary epithelium, J. Clin. Invest. 110 (2002) 815–825. [115] H. Zhao, Y. Cui, J. Dupont, H. Sun, L. Hennighausen, S. Yakar, Overexpression of the tumor suppressor gene phosphatase and tensin homologue partially inhibits wnt-1-induced mammary tumorigenesis, Cancer Res. 65 (2005) 6864–6873. [116] I. Garcia-Cao, M.S. Song, R.M. Hobbs, G. Laurent, C. Giorgi, V.C. de Boer, D. Anastasiou, K. Ito, A.T. Sasaki, L. Rameh, A. Carracedo, M.G. Vander Heiden, L.C. Cantley, P. Pinton, M.C. Haigis, P.P. Pandolfi, Systemic elevation of PTEN induces a tumor-suppressive metabolic state, Cell 149 (2012) 49–62. [117] P. Lopez-Bergami, E. Lau, Z. Ronai, Emerging roles of ATF2 and the dynamic AP1 network in cancer, nature reviews, Cancer 10 (2010) 65–76. [118] M.R. Young, J.J. Li, M. Rincon, R.A. Flavell, B.K. Sathyanarayana, R. Hunziker, N. Colburn, Transgenic mice demonstrate AP-1 (activator protein-1) transactivation is required for tumor promotion, Proc. Natl. Acad. Sci. U. S. A. 96 (1999) 9827–9832. [119] Y. Shimizu, I. Kinoshita, J. Kikuchi, K. Yamazaki, M. Nishimura, M.J. Birrer, H. Dosaka-Akita, Growth inhibition of non-small cell lung cancer cells by AP-1 blockade using a cJun dominant-negative mutant, Br. J. Cancer 98 (2008) 915–922. [120] E. Saez, S.E. Rutberg, E. Mueller, H. Oppenheim, J. Smoluk, S.H. Yuspa, B.M. Spiegelman, c-fos is required for malignant progression of skin tumors, Cell 82 (1995) 721–732. [121] Y. Luo, H. Zhou, M. Mizutani, N. Mizutani, R.A. Reisfeld, R. Xiang, Transcription factor Fos-related antigen 1 is an effective target for a breast cancer vaccine, Proc. Natl. Acad. Sci. U. S. A. 100 (2003) 8850–8855. [122] D.G. Stover, B. Bierie, H.L. Moses, A delicate balance: TGF-beta and the tumor microenvironment, J. Cell. Biochem. 101 (2007) 851–861. [123] A. Joshi, D. Cao, TGF-beta signaling, tumor microenvironment and tumor progression: the butterfly effect, Front. Biosci. 15 (2010) 180–194. [124] D.F. Pierce Jr., A.E. Gorska, A. Chytil, K.S. Meise, D.L. Page, R.J. Coffey Jr., H.L. Moses, Mammary tumor suppression by transforming growth factor beta 1 transgene expression, Proc. Natl. Acad. Sci. U. S. A. 92 (1995) 4254–4258. [125] C.A. Boulanger, G.H. Smith, Reducing mammary cancer risk through premature stem cell senescence, Oncogene 20 (2001) 2264–2272. [126] A. Turco, A. Coppa, S. Aloe, G. Baccheschi, S. Morrone, G. Zupi, G. Colletta, Overexpression of transforming growth factor beta-type II receptor reduces tumorigenicity and metastastic potential of K-ras-transformed thyroid cells, Int. J. Cancer 80 (1999) 85–91. [127] W. Cui, D.J. Fowlis, S. Bryson, E. Duffie, H. Ireland, A. Balmain, R.J. Akhurst, TGFbeta1 inhibits the formation of benign skin tumors, but enhances progression to invasive spindle carcinomas in transgenic mice, Cell 86 (1996) 531–542. [128] K. Matsuzaki, Smad phosphoisoform signaling specificity: the right place at the right time, Carcinogenesis 32 (2011) 1578–1588. [129] A.G. Li, S.L. Lu, M.X. Zhang, C. Deng, X.J. Wang, Smad3 knockout mice exhibit a resistance to skin chemical carcinogenesis, Cancer Res. 64 (2004) 7836–7845. [130] G.S. Ashcroft, X. Yang, A.B. Glick, M. Weinstein, J.L. Letterio, D.E. Mizel, M. Anzano, T. Greenwell-Wild, S.M. Wahl, C. Deng, A.B. Roberts, Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response, Nat. Cell Biol. 1 (1999) 260–266. [131] B. Li, Bone morphogenetic protein-Smad pathway as drug targets for osteoporosis and cancer therapy, Endocr. Metab. Immune Disord. Drug Targets 8 (2008) 208–219. [132] M. Blessing, P. Schirmacher, S. Kaiser, Overexpression of bone morphogenetic protein-6 (BMP-6) in the epidermis of transgenic mice: inhibition or stimulation of proliferation depending on the pattern of transgene expression and formation of psoriatic lesions, J. Cell Biol. 135 (1996) 227–239. [133] S. Wach, P. Schirmacher, M. Protschka, M. Blessing, Overexpression of bone morphogenetic protein-6 (BMP-6) in murine epidermis suppresses skin tumor formation by induction of apoptosis and downregulation of fos/jun family members, Oncogene 20 (2001) 7761–7769. [134] M. Blessing, L.B. Nanney, L.E. King, B.L. Hogan, Chemical skin carcinogenesis is prevented in mice by the induced expression of a TGF-beta related transgene, Teratog. Carcinog. Mutagen. 15 (1995) 11–21. [135] P.N. Kelly, A. Strasser, The role of Bcl-2 and its pro-survival relatives in tumourigenesis and cancer therapy, Cell Death Differ. 18 (2011) 1414–1424. [136] R. Beroukhim, C.H. Mermel, D. Porter, G. Wei, S. Raychaudhuri, J. Donovan, J. Barretina, J.S. Boehm, J. Dobson, M. Urashima, K.T. Mc Henry, R.M. Pinchback, A.H. Ligon, Y.J. Cho, L. Haery, H. Greulich, M. Reich, W. Winckler, M.S. Lawrence, B.A. Weir, K.E. Tanaka, D.Y. Chiang, A.J. Bass, A. Loo, C. Hoffman, J. Prensner, T. Liefeld, Q. Gao, D. Yecies, S. Signoretti, E. Maher, F.J. Kaye, H. Sasaki, J.E. Tepper, J.A. Fletcher, J. Tabernero, J. Baselga, M.S. Tsao, F. Demichelis, M.A. Rubin, P.A. Janne, M.J. Daly, C. Nucera, R.L. Levine, B.L. Ebert, [137] [138] [139] [140] [141] [142] [143] [144] [145] [146] [147] [148] [149] [150] [151] [152] [153] [154] [155] [156] [157] [158] [159] [160] [161] [162] S. Gabriel, A.K. Rustgi, C.R. Antonescu, M. Ladanyi, A. Letai, L.A. Garraway, M. Loda, D.G. Beer, L.D. True, A. Okamoto, S.L. Pomeroy, S. Singer, T.R. Golub, E.S. Lander, G. Getz, W.R. Sellers, M. Meyerson, The landscape of somatic copynumber alteration across human cancers, Nature 463 (2010) 899–905. D.J. Kim, K. Kataoka, S. Sano, K. Connolly, K. Kiguchi, J. DiGiovanni, Targeted disruption of Bcl-xL in mouse keratinocytes inhibits both UVB- and chemically induced skin carcinogenesis, Mol. Carcinog. 48 (2009) 873–885. A. de La Coste, A. Mignon, M. Fabre, E. Gilbert, A. Porteu, T. Van Dyke, A. Kahn, C. Perret, Paradoxical inhibition of c-myc-induced carcinogenesis by Bcl-2 in transgenic mice, Cancer Res. 59 (1999) 5017–5022. R.H. Pierce, M.E. Vail, L. Ralph, J.S. Campbell, N. Fausto, Bcl-2 expression inhibits liver carcinogenesis and delays the development of proliferating foci, Am. J. Pathol. 160 (2002) 1555–1560. H. Rossiter, S. Beissert, C. Mayer, M.P. Schon, B.G. Wienrich, E. Tschachler, T.S. Kupper, Targeted expression of bcl-2 to murine basal epidermal keratinocytes results in paradoxical retardation of ultraviolet- and chemical-induced tumorigenesis, Cancer Res. 61 (2001) 3619–3626. L.M. Fedorov, O.Y. Tyrsin, T. Papadopoulos, G. Camarero, R. Gotz, U.R. Rapp, Bcl-2 determines susceptibility to induction of lung cancer by oncogenic CRaf, Cancer Res. 62 (2002) 6297–6303. J.H. Hao, Q.L. Gu, B.Y. Liu, J.F. Li, X.H. Chen, Y.B. Ji, Z.G. Zhu, Y.Z. Lin, Inhibition of the proliferation of human gastric cancer cells SGC-7901 in vitro and in vivo using Bcl-2 siRNA, Chin. Med. J. 120 (2007) 2105–2111. M. Ilyas, X.P. Hao, K. Wilkinson, I.P. Tomlinson, A.M. Abbasi, A. Forbes, W.F. Bodmer, I.C. Talbot, Loss of Bcl-2 expression correlates with tumour recurrence in colorectal cancer, Gut 43 (1998) 383–387. R.J. Bold, S. Virudachalam, D.J. McConkey, BCL2 expression correlates with metastatic potential in pancreatic cancer cell lines, Cancer 92 (2001) 1122–1129. S.C. Yip, S. Saha, J. Chernoff, PTP1B: a double agent in metabolism and oncogenesis, Trends Biochem. Sci. 35 (2010) 442–449. M. Elchebly, P. Payette, E. Michaliszyn, W. Cromlish, S. Collins, A.L. Loy, D. Normandin, A. Cheng, J. Himms-Hagen, C.C. Chan, C. Ramachandran, M.J. Gresser, M.L. Tremblay, B.P. Kennedy, Increased insulin sensitivity and obesity resistance in mice lacking the protein tyrosine phosphatase-1B gene, Science 283 (1999) 1544–1548. N. Dube, A. Bourdeau, K.M. Heinonen, A. Cheng, A.L. Loy, M.L. Tremblay, Genetic ablation of protein tyrosine phosphatase 1B accelerates lymphomagenesis of p53-null mice through the regulation of B-cell development, Cancer Res. 65 (2005) 10088–10095. S.G. Julien, N. Dube, M. Read, J. Penney, M. Paquet, Y. Han, B.P. Kennedy, W.J. Muller, M.L. Tremblay, Protein tyrosine phosphatase 1B deficiency or inhibition delays ErbB2-induced mammary tumorigenesis and protects from lung metastasis, Nat. Genet. 39 (2007) 338–346. M. Bentires-Alj, B.G. Neel, Protein-tyrosine phosphatase 1B is required for HER2/Neu-induced breast cancer, Cancer Res. 67 (2007) 2420–2424. Y. Pylayeva-Gupta, E. Grabocka, D. Bar-Sagi, RAS oncogenes: weaving a tumorigenic web, nature reviews, Cancer 11 (2011) 761–774. L. van der Weyden, D.J. Adams, The Ras-association domain family (RASSF) members and their role in human tumourigenesis, Biochim. Biophys. Acta 1776 (2007) 58–85. I.C. Jacquemart, A.E. Springs, W.Y. Chen, Rassf3 is responsible in part for resistance to mammary tumor development in neu transgenic mice, Int. J. Oncol. 34 (2009) 517–528. R. Bosco, E. Melloni, C. Celeghini, E. Rimondi, M. Vaccarezza, G. Zauli, Fine tuning of protein kinase C (PKC) isoforms in cancer: shortening the distance from the laboratory to the bedside, Mini Rev. Med. Chem. 11 (2011) 185–199. H. Mischak, J.A. Goodnight, W. Kolch, G. Martiny-Baron, C. Schaechtle, M.G. Kazanietz, P.M. Blumberg, J.H. Pierce, J.F. Mushinski, Overexpression of protein kinase C-delta and -epsilon in NIH 3T3 cells induces opposite effects on growth, morphology, anchorage dependence, and tumorigenicity, J. Biol. Chem. 268 (1993) 6090–6096. P.J. Reddig, N.E. Dreckschmidt, H. Ahrens, R. Simsiman, C.P. Tseng, J. Zou, T.D. Oberley, A.K. Verma, Transgenic mice overexpressing protein kinase Cdelta in the epidermis are resistant to skin tumor promotion by 12-O-tetradecanoylphorbol13-acetate, Cancer Res. 59 (1999) 5710–5718. L.V. Mauro, V.C. Grossoni, A.J. Urtreger, C. Yang, L.L. Colombo, A. Morandi, M.G. Pallotta, M.G. Kazanietz, E.D. Bal de Kier Joffe, L.L. Puricelli, PKC Delta (PKCdelta) promotes tumoral progression of human ductal pancreatic cancer, Pancreas 39 (2010) e31–e41. J.A. Lowry, W.R. Atchley, Molecular evolution of the GATA family of transcription factors: conservation within the DNA-binding domain, J. Mol. Evol. 50 (2000) 103–115. R. Zheng, G.A. Blobel, GATA transcription factors and cancer, Genes Cancer 1 (2010) 1178–1188. D. Kamnasaran, B. Qian, C. Hawkins, W.L. Stanford, A. Guha, GATA6 is an astrocytoma tumor suppressor gene identified by gene trapping of mouse glioma model, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 8053–8058. S. Agnihotri, A. Wolf, D. Picard, C. Hawkins, A. Guha, GATA4 is a regulator of astrocyte cell proliferation and apoptosis in the human and murine central nervous system, Oncogene 28 (2009) 3033–3046. S. Agnihotri, A. Wolf, D.M. Munoz, C.J. Smith, A. Gajadhar, A. Restrepo, I.D. Clarke, G.N. Fuller, S. Kesari, P.B. Dirks, C.J. McGlade, W.L. Stanford, K. Aldape, P.S. Mischel, C. Hawkins, A. Guha, A GATA4-regulated tumor suppressor network represses formation of malignant human astrocytomas, J. Exp. Med. 208 (2011) 689–702. S.F. Sells, D.P. Wood Jr., S.S. Joshi-Barve, S. Muthukumar, R.J. Jacob, S.A. Crist, S. Humphreys, V.M. Rangnekar, Commonality of the gene programs induced by L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 [163] [164] [165] [166] [167] [168] [169] [170] [171] [172] [173] [174] [175] [176] [177] [178] [179] [180] [181] [182] [183] [184] [185] [186] effectors of apoptosis in androgen-dependent and -independent prostate cells, Cell Growth Differ. 5 (1994) 457–466. T. Shrestha-Bhattarai, V.M. Rangnekar, Cancer-selective apoptotic effects of extracellular and intracellular Par-4, Oncogene 29 (2010) 3873–3880. M. Chakraborty, S.G. Qiu, K.M. Vasudevan, V.M. Rangnekar, Par-4 drives trafficking and activation of Fas and Fasl to induce prostate cancer cell apoptosis and tumor regression, Cancer Res. 61 (2001) 7255–7263. T. Lucas, B. Pratscher, S. Krishnan, D. Fink, P. Gunsberg, M. Wolschek, V. Wacheck, T. Muster, I. Romirer, K. Wolff, H. Pehamberger, H.G. Eichler, V.M. Rangnekar, B. Jansen, Differential expression levels of Par-4 in melanoma, Melanoma Res. 11 (2001) 379–383. Y. Zhao, R. Burikhanov, S. Qiu, S.M. Lele, C.D. Jennings, S. Bondada, B. Spear, V.M. Rangnekar, Cancer resistance in transgenic mice expressing the SAC module of Par-4, Cancer Res. 67 (2007) 9276–9285. Y. Zhao, R. Burikhanov, J. Brandon, S. Qiu, B.J. Shelton, B. Spear, S. Bondada, S. Bryson, V.M. Rangnekar, Systemic Par-4 inhibits non-autochthonous tumor growth, Cancer Biol. Ther. 12 (2011) 152–157. R. Burikhanov, Y. Zhao, A. Goswami, S. Qiu, S.R. Schwarze, V.M. Rangnekar, The tumor suppressor Par-4 activates an extrinsic pathway for apoptosis, Cell 138 (2009) 377–388. X. Lu, B. Nannenga, L.A. Donehower, PPM1D dephosphorylates Chk1 and p53 and abrogates cell cycle checkpoints, Genes Dev. 19 (2005) 1162–1174. D.V. Bulavin, C. Phillips, B. Nannenga, O. Timofeev, L.A. Donehower, C.W. Anderson, E. Appella, A.J. Fornace Jr., Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16(Ink4a)-p19(Arf) pathway, Nat. Genet. 36 (2004) 343–350. H. Fujimoto, N. Onishi, N. Kato, M. Takekawa, X.Z. Xu, A. Kosugi, T. Kondo, M. Imamura, I. Oishi, A. Yoda, Y. Minami, Regulation of the antioncogenic Chk2 kinase by the oncogenic Wip1 phosphatase, Cell Death Differ. 13 (2006) 1170–1180. X. Lu, T.A. Nguyen, S.H. Moon, Y. Darlington, M. Sommer, L.A. Donehower, The type 2C phosphatase Wip1: an oncogenic regulator of tumor suppressor and DNA damage response pathways, Cancer Metastasis Rev. 27 (2008) 123–135. D.S. Tan, M.B. Lambros, S. Rayter, R. Natrajan, R. Vatcheva, Q. Gao, C. Marchio, F.C. Geyer, K. Savage, S. Parry, K. Fenwick, N. Tamber, A. Mackay, T. Dexter, C. Jameson, W.G. McCluggage, A. Williams, A. Graham, D. Faratian, M. ElBahrawy, A.J. Paige, H. Gabra, M.E. Gore, M. Zvelebil, C.J. Lord, S.B. Kaye, A. Ashworth, J.S. Reis-Filho, PPM1D is a potential therapeutic target in ovarian clear cell carcinomas, Clin. Cancer Res. 15 (2009) 2269–2280. B. Nannenga, X. Lu, M. Dumble, M. Van Maanen, T.A. Nguyen, R. Sutton, T.R. Kumar, L.A. Donehower, Augmented cancer resistance and DNA damage response phenotypes in PPM1D null mice, Mol. Carcinog. 45 (2006) 594–604. G.I. Belova, O.N. Demidov, A.J. Fornace Jr., D.V. Bulavin, Chemical inhibition of Wip1 phosphatase contributes to suppression of tumorigenesis, Cancer Biol. Ther. 4 (2005) 1154–1158. J. Yelamos, J. Farres, L. Llacuna, C. Ampurdanes, J. Martin-Caballero, PARP-1 and PARP-2: new players in tumour development, Am. J. Cancer Res. 1 (2011) 328–346. M. Tsutsumi, M. Masutani, T. Nozaki, O. Kusuoka, T. Tsujiuchi, H. Nakagama, H. Suzuki, Y. Konishi, T. Sugimura, Increased susceptibility of poly(ADP-ribose) polymerase-1 knockout mice to nitrosamine carcinogenicity, Carcinogenesis 22 (2001) 1–3. T. Nozaki, H. Fujihara, M. Watanabe, M. Tsutsumi, K. Nakamoto, O. Kusuoka, N. Kamada, H. Suzuki, H. Nakagama, T. Sugimura, M. Masutani, Parp-1 deficiency implicated in colon and liver tumorigenesis induced by azoxymethane, Cancer Sci. 94 (2003) 497–500. A. Gunji, A. Uemura, M. Tsutsumi, T. Nozaki, O. Kusuoka, K. Omura, H. Suzuki, H. Nakagama, T. Sugimura, M. Masutani, Parp-1 deficiency does not increase the frequency of tumors in the oral cavity and esophagus of ICR/129Sv mice by 4nitroquinoline 1-oxide, a carcinogen producing bulky adducts, Cancer Lett. 241 (2006) 87–92. D. Martin-Oliva, F. O'Valle, J.A. Munoz-Gamez, M.T. Valenzuela, M.I. Nunez, M. Aguilar, J.M. Ruiz de Almodovar, R. Garcia del Moral, F.J. Oliver, Crosstalk between PARP-1 and NF-kappaB modulates the promotion of skin neoplasia, Oncogene 23 (2004) 5275–5283. D. Martin-Oliva, R. Aguilar-Quesada, F. O'Valle, J.A. Munoz-Gamez, R. MartinezRomero, R. Garcia Del Moral, J.M. Ruiz de Almodovar, R. Villuendas, M.A. Piris, F.J. Oliver, Inhibition of poly(ADP-ribose) polymerase modulates tumor-related gene expression, including hypoxia-inducible factor-1 activation, during skin carcinogenesis, Cancer Res. 66 (2006) 5744–5756. C. Conde, M. Mark, F.J. Oliver, A. Huber, G. de Murcia, J. Menissier-de Murcia, Loss of poly(ADP-ribose) polymerase-1 causes increased tumour latency in p53deficient mice, EMBO J. 20 (2001) 3535–3543. R. Beneke, T. Moroy, Inhibition of poly(ADP-ribose) polymerase activity accelerates T-cell lymphomagenesis in p53 deficient mice, Oncogene 20 (2001) 8136–8141. W.M. Tong, H. Ohgaki, H. Huang, C. Granier, P. Kleihues, Z.Q. Wang, Null mutation of DNA strand break-binding molecule poly(ADP-ribose) polymerase causes medulloblastomas in p53(−/−) mice, Am. J. Pathol. 162 (2003) 343–352. W.M. Tong, M.P. Hande, P.M. Lansdorp, Z.Q. Wang, DNA strand break-sensing molecule poly(ADP-Ribose) polymerase cooperates with p53 in telomere function, chromosome stability, and tumor suppression, Mol. Cell. Biol. 21 (2001) 4046–4054. W.M. Tong, U. Cortes, M.P. Hande, H. Ohgaki, L.R. Cavalli, P.M. Lansdorp, B.R. Haddad, Z.Q. Wang, Synergistic role of Ku80 and poly(ADP-ribose) polymerase in suppressing chromosomal aberrations and liver cancer formation, Cancer Res. 62 (2002) 6990–6996. 329 [187] M. Lebel, J. Lavoie, I. Gaudreault, M. Bronsard, R. Drouin, Genetic cooperation between the Werner syndrome protein and poly(ADP-ribose) polymerase-1 in preventing chromatid breaks, complex chromosomal rearrangements, and cancer in mice, Am. J. Pathol. 162 (2003) 1559–1569. [188] L. Tentori, A. Muzi, A.S. Dorio, S. Bultrini, E. Mazzon, P.M. Lacal, G.M. Shah, J. Zhang, P. Navarra, G. Nocentini, S. Cuzzocrea, G. Graziani, Stable depletion of poly (ADP-ribose) polymerase-1 reduces in vivo melanoma growth and increases chemosensitivity, Eur. J. Cancer 44 (2008) 1302–1314. [189] H.E. Bryant, N. Schultz, H.D. Thomas, K.M. Parker, D. Flower, E. Lopez, S. Kyle, M. Meuth, N.J. Curtin, T. Helleday, Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase, Nature 434 (2005) 913–917. [190] P.C. Fong, D.S. Boss, T.A. Yap, A. Tutt, P. Wu, M. Mergui-Roelvink, P. Mortimer, H. Swaisland, A. Lau, M.J. O'Connor, A. Ashworth, J. Carmichael, S.B. Kaye, J.H. Schellens, J.S. de Bono, Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers, N. Engl. J. Med. 361 (2009) 123–134. [191] M. Rouleau, A. Patel, M.J. Hendzel, S.H. Kaufmann, G.G. Poirier, PARP inhibition: PARP1 and beyond, nature reviews, Cancer 10 (2010) 293–301. [192] M. Christmann, B. Verbeek, W.P. Roos, B. Kaina, O(6)-Methylguanine-DNA methyltransferase (MGMT) in normal tissues and tumors: enzyme activity, promoter methylation and immunohistochemistry, Biochim. Biophys. Acta 1816 (2011) 179–190. [193] L.L. Dumenco, E. Allay, K. Norton, S.L. Gerson, The prevention of thymic lymphomas in transgenic mice by human O6-alkylguanine-DNA alkyltransferase, Science 259 (1993) 219–222. [194] L. Liu, E. Allay, L.L. Dumenco, S.L. Gerson, Rapid repair of O6-methylguanineDNA adducts protects transgenic mice from N-methylnitrosourea-induced thymic lymphomas, Cancer Res. 54 (1994) 4648–4652. [195] N.H. Zaidi, E. Allay, T.C. Ayi, B.F. Li, L.L. Dumenco, M.S. Sy, S.L. Gerson, The immature thymocyte is protected from N-methylnitrosourea-induced lymphoma by the human MGMT-CD2 transgene, Carcinogenesis 16 (1995) 1047–1053. [196] E. Allay, M. Veigl, S.L. Gerson, Mice over-expressing human O6 alkylguanineDNA alkyltransferase selectively reduce O6 methylguanine mediated carcinogenic mutations to threshold levels after N-methyl-N-nitrosourea, Oncogene 18 (1999) 3783–3787. [197] N.H. Zaidi, T.P. Pretlow, M.A. O'Riordan, L.L. Dumenco, E. Allay, S.L. Gerson, Transgenic expression of human MGMT protects against azoxymethaneinduced aberrant crypt foci and G to A mutations in the K-ras oncogene of mouse colon, Carcinogenesis 16 (1995) 451–456. [198] L. Liu, X. Qin, S.L. Gerson, Reduced lung tumorigenesis in human methylguanine DNA—methyltransferase transgenic mice achieved by expression of transgene within the target cell, Carcinogenesis 20 (1999) 279–284. [199] K. Becker, C. Gregel, C. Fricke, D. Komitowski, J. Dosch, B. Kaina, DNA repair protein MGMT protects against N-methyl-N-nitrosourea-induced conversion of benign into malignant tumors, Carcinogenesis 24 (2003) 541–546. [200] Y. Nakatsuru, S. Matsukuma, N. Nemoto, H. Sugano, M. Sekiguchi, T. Ishikawa, O6-methylguanine-DNA methyltransferase protects against nitrosamine-induced hepatocarcinogenesis, Proc. Natl. Acad. Sci. U. S. A. 90 (1993) 6468–6472. [201] X. Qin, S. Zhang, S. Matsukuma, M. Zarkovic, S. Shimizu, T. Ishikawa, Y. Nakatsuru, Protection against malignant progression of spontaneously developing liver tumors in transgenic mice expressing O(6)-methylguanine-DNA methyltransferase, Jpn. J. Cancer Res. 91 (2000) 1085–1089. [202] X. Qin, H. Zhou, L. Liu, S.L. Gerson, Transgenic expression of human MGMT blocks the hypersensitivity of PMS2-deficient mice to low dose MNU thymic lymphomagenesis, Carcinogenesis 20 (1999) 1667–1673. [203] J.S. Reese, E. Allay, S.L. Gerson, Overexpression of human O6-alkylguanine DNA alkyltransferase (AGT) prevents MNU induced lymphomas in heterozygous p53 deficient mice, Oncogene 20 (2001) 5258–5263. [204] E. Allay, J.S. Reese, E.A. McGuire, O.N. Koc, N. Sedransk, S.L. Gerson, Potentiation of lymphomagenesis by methylnitrosourea in mice transgenic for LMO1 is blocked by O6-alkylguanine DNA-alkyltransferase, Oncogene 15 (1997) 2127–2132. [205] Z.Q. Zhou, D. Manguino, K. Kewitt, G.W. Intano, C.A. McMahan, D.C. Herbert, M. Hanes, R. Reddick, Y. Ikeno, C.A. Walter, Spontaneous hepatocellular carcinoma is reduced in transgenic mice overexpressing human O6- methylguanine-DNA methyltransferase, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 12566–12571. [206] F.I. Daniel, K. Cherubini, L.S. Yurgel, M.A. de Figueiredo, F.G. Salum, The role of epigenetic transcription repression and DNA methyltransferases in cancer, Cancer 117 (2011) 677–687. [207] R.T. Cormier, W.F. Dove, Dnmt1N/+ reduces the net growth rate and multiplicity of intestinal adenomas in C57BL/6-multiple intestinal neoplasia (Min)/+ mice independently of p53 but demonstrates strong synergy with the modifier of Min 1(AKR) resistance allele, Cancer Res. 60 (2000) 3965–3970. [208] P.W. Laird, L. Jackson-Grusby, A. Fazeli, S.L. Dickinson, W.E. Jung, E. Li, R.A. Weinberg, R. Jaenisch, Suppression of intestinal neoplasia by DNA hypomethylation, Cell 81 (1995) 197–205. [209] C.A. Eads, A.E. Nickel, P.W. Laird, Complete genetic suppression of polyp formation and reduction of CpG-island hypermethylation in Apc(Min/+) Dnmt1hypomorphic mice, Cancer Res. 62 (2002) 1296–1299. [210] B.N. Trinh, T.I. Long, A.E. Nickel, D. Shibata, P.W. Laird, DNA methyltransferase deficiency modifies cancer susceptibility in mice lacking DNA mismatch repair, Mol. Cell. Biol. 22 (2002) 2906–2917. [211] S. Oghamian, N.M. Sodir, M.U. Bashir, H. Shen, A.E. Cullins, C.A. Carroll, P. Kundu, D. Shibata, P.W. Laird, Reduction of pancreatic acinar cell tumor multiplicity in Dnmt1 hypomorphic mice, Carcinogenesis 32 (2011) 829–835. [212] Y. Cao, T.M. Bryan, R.R. Reddel, Increased copy number of the TERT and TERC telomerase subunit genes in cancer cells, Cancer Sci. 99 (2008) 1092–1099. 330 L. van der Weyden, D.J. Adams / Biochimica et Biophysica Acta 1826 (2012) 312–330 [213] E. Gonzalez-Suarez, E. Samper, J.M. Flores, M.A. Blasco, Telomerase-deficient mice with short telomeres are resistant to skin tumorigenesis, Nat. Genet. 26 (2000) 114–117. [214] P.A. Farazi, J. Glickman, S. Jiang, A. Yu, K.L. Rudolph, R.A. DePinho, Differential impact of telomere dysfunction on initiation and progression of hepatocellular carcinoma, Cancer Res. 63 (2003) 5021–5027. [215] R.A. Greenberg, L. Chin, A. Femino, K.H. Lee, G.J. Gottlieb, R.H. Singer, C.W. Greider, R.A. DePinho, Short dysfunctional telomeres impair tumorigenesis in the INK4a(delta2/3) cancer-prone mouse, Cell 97 (1999) 515–525. [216] K.L. Rudolph, M. Millard, M.W. Bosenberg, R.A. DePinho, Telomere dysfunction and evolution of intestinal carcinoma in mice and humans, Nat. Genet. 28 (2001) 155–159. [217] L. Qi, M.A. Strong, B.O. Karim, D.L. Huso, C.W. Greider, Telomere fusion to chromosome breaks reduces oncogenic translocations and tumour formation, Nat. Cell Biol. 7 (2005) 706–711. [218] W. Cosme-Blanco, M.F. Shen, A.J. Lazar, S. Pathak, G. Lozano, A.S. Multani, S. Chang, Telomere dysfunction suppresses spontaneous tumorigenesis in vivo by initiating p53-dependent cellular senescence, EMBO Rep. 8 (2007) 497–503. [219] M. Jaskelioff, W. Song, J. Xia, C. Liu, J. Kramer, S. Koido, S.J. Gendler, S.K. Calderwood, J. Gong, Telomerase deficiency and telomere dysfunction inhibit mammary tumors induced by polyomavirus middle T oncogene, Oncogene 28 (2009) 4225–4236. [220] J.Y. Liu, Q. Zhu, J. Li, S. Zhao, L. Li, The retrovirus-mediated antisense human telomerase RNA (hTR) gene limits the growth of hepatocellular carcinoma growth in cell culture and animals, Dig. Dis. Sci. 53 (2008) 1122–1130. [221] S.M. Yang, D.C. Fang, J.L. Yang, L. Chen, Y.H. Luo, G.P. Liang, Antisense human telomerase reverse transcriptase could partially reverse malignant phenotypes of gastric carcinoma cell line in vitro, Eur. J. Cancer Prev. 17 (2008) 209–217. [222] F. Noreen, J. Heinrich, K. Moelling, Antitumor activity of small double-stranded oligodeoxynucleotides targeting telomerase RNA in malignant melanoma cells, Oligonucleotides 19 (2009) 169–178. [223] M. Nosrati, S. Li, S. Bagheri, D. Ginzinger, E.H. Blackburn, R.J. Debs, M. KashaniSabet, Antitumor activity of systemically delivered ribozymes targeting murine telomerase RNA, Clin. Cancer Res. 10 (2004) 4983–4990. [224] B. Bojovic, D.L. Crowe, Telomere dysfunction promotes metastasis in a TERC null mouse model of head and neck cancer, Mol. Cancer Res. 9 (2011) 901–913. [225] J. Walker-Daniels, A.R. Hess, M.J. Hendrix, M.S. Kinch, Differential regulation of EphA2 in normal and malignant cells, Am. J. Pathol. 162 (2003) 1037–1042. [226] W. Yuan, Z. Chen, Z. Chen, S. Wu, J. Guo, J. Ge, P. Yang, J. Huang, Silencing of EphA2 inhibits invasion of human gastric cancer SGC-7901 cells in vitro and in vivo, Neoplasma 59 (2012) 105–113. [227] W.B. Fang, D.M. Brantley-Sieders, M.A. Parker, A.D. Reith, J. Chen, A kinasedependent role for EphA2 receptor in promoting tumor growth and metastasis, Oncogene 24 (2005) 7859–7868. [228] D.M. Brantley-Sieders, G. Zhuang, D. Hicks, W.B. Fang, Y. Hwang, J.M. Cates, K. Coffman, D. Jackson, E. Bruckheimer, R.S. Muraoka-Cook, J. Chen, The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling, J. Clin. Invest. 118 (2008) 64–78. [229] C. Bogan, J. Chen, M.G. O'Sullivan, R.T. Cormier, Loss of EphA2 receptor tyrosine kinase reduces ApcMin/+ tumorigenesis, Int. J. Cancer 124 (2009) 1366–1371. [230] H. Guo, H. Miao, L. Gerber, J. Singh, M.F. Denning, A.C. Gilliam, B. Wang, Disruption of EphA2 receptor tyrosine kinase leads to increased susceptibility to carcinogenesis in mouse skin, Cancer Res. 66 (2006) 7050–7058. [231] S. Abuharbeid, F. Czubayko, A. Aigner, The fibroblast growth factor-binding protein FGF-BP, Int. J. Biochem. Cell Biol. 38 (2006) 1463–1468. [232] F. Czubayko, E.D. Liaudet-Coopman, A. Aigner, A.T. Tuveson, G.J. Berchem, A. Wellstein, A secreted FGF-binding protein can serve as the angiogenic switch in human cancer, Nat. Med. 3 (1997) 1137–1140. [233] D. Schulze, P. Plohmann, S. Hobel, A. Aigner, Anti-tumor effects of fibroblast growth factor-binding protein (FGF-BP) knockdown in colon carcinoma, Mol. Cancer 10 (2011) 144. [234] Z.A. Sherif, S. Nakai, K.F. Pirollo, A. Rait, E.H. Chang, Downmodulation of bFGFbinding protein expression following restoration of p53 function, Cancer Gene Ther. 8 (2001) 771–782. [235] E.D. Liaudet-Coopman, G.J. Berchem, A. Wellstein, In vivo inhibition of angiogenesis and induction of apoptosis by retinoic acid in squamous cell carcinoma, Clin. Cancer Res. 3 (1997) 179–184. [236] H. Nagase, S. Bryson, H. Cordell, C.J. Kemp, F. Fee, A. Balmain, Distinct genetic loci control development of benign and malignant skin tumours in mice, Nat. Genet. 10 (1995) 424–429. [237] G. Manenti, M. Gariboldi, R. Elango, A. Fiorino, L. De Gregorio, F.S. Falvella, K. Hunter, D. Housman, M.A. Pierotti, T.A. Dragani, Genetic mapping of a pulmonary adenoma resistance (Par1) in mouse, Nat. Genet. 12 (1996) 455–457. [238] M. Wang, Z. Zhang, Z. Zhang, H. Vikis, Y. Yan, Y. Wang, M. You, Fine mapping and candidate gene analyses of pulmonary adenoma resistance 1, a major genetic determinant of mouse lung adenoma resistance, Cancer Res. 67 (2007) 2508–2516. [239] A. Pataer, M. Nishimura, T. Kamoto, K. Ichioka, M. Sato, H. Hiai, Genetic resistance to urethan-induced pulmonary adenomas in SMXA recombinant inbred mouse strains, Cancer Res. 57 (1997) 2904–2908. [240] H. Hiai, P. Abujiang, M. Nishimura, Polygenic resistance to mouse pulmonary adenomas, Exp. Lung Res. 26 (2000) 617–625. [241] M. Obata, H. Nishimori, K. Ogawa, G.H. Lee, Identification of the Par2 (pulmonary adenoma resistance) locus on mouse chromosome 18, a major genetic determinant for lung carcinogen resistance in BALB/cByJ mice, Oncogene 13 (1996) 1599–1604. [242] Z. Zhang, L. Lin, G. Liu, M. Wang, J. Hill, Y. Wang, M. You, T.R. Devereux, Fine mapping and characterization of candidate lung tumor resistance genes for the Par2 locus on mouse chromosome 18, Exp. Lung Res. 26 (2000) 627–639. [243] G.H. Lee, H. Matsushita, T. Kitagawa, Fine chromosomal localization of the mouse Par2 gene that confers resistance against urethane-induction of pulmonary adenomas, Oncogene 20 (2001) 3979–3985. [244] X. Qin, L. Liu, S.L. Gerson, Mice defective in the DNA mismatch gene PMS2 are hypersensitive to MNU induced thymic lymphoma and are partially protected by transgenic expression of human MGMT, Oncogene 18 (1999) 4394–4400. [245] R.J. Sharpe, H.R. Byers, C.F. Scott, S.I. Bauer, T.E. Maione, Growth inhibition of murine melanoma and human colon carcinoma by recombinant human platelet factor 4, J. Natl. Cancer Inst. 82 (1990) 848–853. [246] D.L. Kolber, T.L. Knisely, T.E. Maione, Inhibition of development of murine melanoma lung metastases by systemic administration of recombinant platelet factor 4, J. Natl. Cancer Inst. 87 (1995) 304–309. [247] S. Hillinger, S.C. Yang, L. Zhu, M. Huang, R. Duckett, K. Atianzar, R.K. Batra, R.M. Strieter, S.M. Dubinett, S. Sharma, EBV-induced molecule 1 ligand chemokine (ELC/CCL19) promotes IFN-gamma-dependent antitumor responses in a lung cancer model, J. Immunol. 171 (2003) 6457–6465. [248] S. Hillinger, S.C. Yang, R.K. Batra, R.M. Strieter, W. Weder, S.M. Dubinett, S. Sharma, CCL19 reduces tumour burden in a model of advanced lung cancer, Br. J. Cancer 94 (2006) 1029–1034. [249] M.P. Colombo, M. Vagliani, F. Spreafico, M. Parenza, C. Chiodoni, C. Melani, A. Stoppacciaro, Amount of interleukin 12 available at the tumor site is critical for tumor regression, Cancer Res. 56 (1996) 2531–2534. [250] Y. Yoshida, K. Tasaki, M. Miyauchi, M. Narita, K. Takenaga, H. Yamamoto, T. Yaaguchi, H. Saisho, S. Sakiyama, M. Tagawa, Impaired tumorigenicity of human pancreatic cancer cells retrovirally transduced with interleukin-12 or interleukin-15 gene, Cancer Gene Ther. 7 (2000) 324–331. [251] K. Tasaki, Y. Yoshida, M. Miyauchi, T. Maeda, K. Takenaga, T. Kouzu, T. Asano, T. Ochiai, S. Sakiyamna, M. Tagawa, Transduction of murine colon carcinoma cells with interleukin-15 gene induces antitumor effects in immunocompetent and immunocompromised hosts, Cancer Gene Ther. 7 (2000) 255–261. [252] B.W. Tse, P.J. Russell, M. Lochner, I. Forster, C.A. Power, IL-18 inhibits growth of murine orthotopic prostate carcinomas via both adaptive and innate immune mechanisms, PLoS One 6 (2011) e24241. [253] B. Shan, L. Yu, O. Shimozato, Q. Li, M. Tagawa, Expression of interleukin-21 and ‐ 23 in human esophageal tumors produced antitumor effects in nude mice, Anticancer Res. 24 (2004) 79–82. [254] Y.Q. Wang, S. Ugai, O. Shimozato, L. Yu, K. Kawamura, H. Yamamoto, T. Yamaguchi, H. Saisho, M. Tagawa, Induction of systemic immunity by expression of interleukin-23 in murine colon carcinoma cells, Int. J. Cancer 105 (2003) 820–824. [255] C.H. Lo, S.C. Lee, P.Y. Wu, W.Y. Pan, J. Su, C.W. Cheng, S.R. Roffler, B.L. Chiang, C.N. Lee, C.W. Wu, M.H. Tao, Antitumor and antimetastatic activity of IL-23, J. Immunol. 171 (2003) 600–607. [256] R. Salcedo, J.K. Stauffer, E. Lincoln, T.C. Back, J.A. Hixon, C. Hahn, K. ShaferWeaver, A. Malyguine, R. Kastelein, J.M. Wigginton, IL-27 mediates complete regression of orthotopic primary and metastatic murine neuroblastoma tumors: role for CD8 + T cells, J. Immunol. 173 (2004) 7170–7182. [257] S.E. Braun, K. Chen, R.G. Foster, C.H. Kim, R. Hromas, M.H. Kaplan, H.E. Broxmeyer, K. Cornetta, The CC chemokine CK beta-11/MIP-3 beta/ELC/Exodus 3 mediates tumor rejection of murine breast cancer cells through NK cells, J. Immunol. 164 (2000) 4025–4031. [258] S.C. Yang, S. Hillinger, K. Riedl, L. Zhang, L. Zhu, M. Huang, K. Atianzar, B.Y. Kuo, B. Gardner, R.K. Batra, R.M. Strieter, S.M. Dubinett, S. Sharma, Intratumoral administration of dendritic cells overexpressing CCL21 generates systemic antitumor responses and confers tumor immunity, Clin. Cancer Res. 10 (2004) 2891–2901. [259] S.C. Yang, R.K. Batra, S. Hillinger, K.L. Reckamp, R.M. Strieter, S.M. Dubinett, S. Sharma, Intrapulmonary administration of CCL21 gene-modified dendritic cells reduces tumor burden in spontaneous murine bronchoalveolar cell carcinoma, Cancer Res. 66 (2006) 3205–3213. [260] Y. Li, Y. Jin, H. Chen, G. Jie, G. Tobelem, J.P. Caen, Z.C. Han, Suppression of tumor growth by viral vector-mediated gene transfer of N-terminal truncated platelet factor 4, Cancer Biother. Radiopharm. 18 (2003) 829–840. [261] K. Yamaguchi, K. Ogawa, T. Katsube, K. Shimao, S. Konno, T. Shimakawa, K. Yoshimatsu, Y. Naritaka, H. Yagawa, K. Hirose, Platelet factor 4 gene transfection into tumor cells inhibits angiogenesis, tumor growth and metastasis, Anticancer Res. 25 (2005) 847–851. [262] L. Yang, J. Du, J. Hou, H. Jiang, J. Zou, Platelet factor-4 and its p17–70 peptide inhibit myeloma proliferation and angiogenesis in vivo, BMC Cancer 11 (2011) 261.