Browse > Article
http://dx.doi.org/10.5483/BMBRep.2014.47.2.005

Exploiting tumor cell senescence in anticancer therapy  

Lee, Minyoung (Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences)
Lee, Jae-Seon (Research Center for Radio-senescence, Korea Institute of Radiological and Medical Sciences)
Publication Information
BMB Reports / v.47, no.2, 2014 , pp. 51-59 More about this Journal
Abstract
Cellular senescence is a physiological process of irreversible cell-cycle arrest that contributes to various physiological and pathological processes of aging. Whereas replicative senescence is associated with telomere attrition after repeated cell division, stress-induced premature senescence occurs in response to aberrant oncogenic signaling, oxidative stress, and DNA damage which is independent of telomere dysfunction. Recent evidence indicates that cellular senescence provides a barrier to tumorigenesis and is a determinant of the outcome of cancer treatment. However, the senescence-associated secretory phenotype, which contributes to multiple facets of senescent cancer cells, may influence both cancer-inhibitory and cancer-promoting mechanisms of neighboring cells. Conventional treatments, such as chemo- and radiotherapies, preferentially induce premature senescence instead of apoptosis in the appropriate cellular context. In addition, treatment-induced premature senescence could compensate for resistance to apoptosis via alternative signaling pathways. Therefore, we believe that an intensive effort to understand cancer cell senescence could facilitate the development of novel therapeutic strategies for improving the efficacy of anticancer therapies. This review summarizes the current understanding of molecular mechanisms, functions, and clinical applications of cellular senescence for anticancer therapy.
Keywords
Anticancer therapy; Cellular senescence; Tumorigenesis; SASP;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Wadhwa, R., Sugihara, T., Taira, K. and Kaul, S. C. (2004) The ARF-p53 senescence pathway in mouse and human cells. Histol. Histopathol. 19, 311-316.
2 Parrinello, S., Coppe, J. P., Krtolica, A. and Campisi, J. (2005) Stromal-epithelial interactions in aging and cancer: senescent fibroblasts alter epithelial cell differentiation. J. Cell Sci. 118, 485-496.   DOI   ScienceOn
3 Salminen, A., Kauppinen, A. and Kaarniranta, K. (2012) Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal. 24, 835-845.   DOI   ScienceOn
4 Coppe, J. P., Kauser, K., Campisi, J. and Beausejour, C. M. (2006) Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J. Biol. Chem. 281, 29568-29574.   DOI   ScienceOn
5 Gosselin, K., Martien, S., Pourtier, A., Vercamer, C., Ostoich, P., Morat, L., Sabatier, L., Duprez, L., T'Kint de Roodenbeke, C., Gilson, E., Malaquin, N., Wernert, N., Slijepcevic, P., Ashtari, M., Chelli, F., Deruy, E., Vandenbunder, B., De Launoit, Y. and Abbadie, C. (2009) Senescence-associated oxidative DNA damage promotes the generation of neoplastic cells. Cancer Res. 69, 7917-7925.   DOI   ScienceOn
6 Acosta, J. C., O'Loghlen, A., Banito, A., Raguz, S. and Gil, J. (2008) Control of senescence by CXCR2 and its ligands. Cell Cycle 7, 2956-2959.   DOI
7 Bavik, C., Coleman, I., Dean, J. P., Knudsen, B., Plymate, S. and Nelson, P. S. (2006) The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res. 66, 794-802.   DOI   ScienceOn
8 Sagiv, A. and Krizhanovsky, V. (2013) Immunosurveillance of senescent cells: the bright side of the senescence program. Biogerontology. 14, 617-628.   DOI   ScienceOn
9 Mariotti, M., Castiglioni, S., Bernardini, D. and Maier, J. A. (2006) Interleukin 1 alpha is a marker of endothelial cellular senescent. Immun. Ageing 3, 4.   DOI   ScienceOn
10 Orjalo, A. V., Bhaumik, D., Gengler, B. K., Scott, G. K. and Campisi, J. (2009) Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proc. Natl. Acad. Sci. U. S. A. 106, 17031-17036.   DOI   ScienceOn
11 Novakova, Z., Hubackova, S., Kosar, M., Janderova- Rossmeislova, L., Dobrovolna, J., Vasicova, P., Vancurova, M., Horejsi, Z., Hozak, P., Bartek, J. and Hodny, Z. (2010) Cytokine expression and signaling in drug-induced cellular senescence. Oncogene 29, 273-284.   DOI   ScienceOn
12 Coppe, J. P., Patil, C. K., Rodier, F., Sun, Y., Munoz, D. P., Goldstein, J., Nelson, P. S., Desprez, P. Y. and Campisi, J. (2008) Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853-2868.
13 Puche, J. E. and Castilla-Cortazar, I. (2012) Human conditions of insulin-like growth factor-I (IGF-I) deficiency. J. Transl. Med. 10, 224.   DOI
14 Lopez-Bermejo, A., Buckway, C. K., Devi, G. R., Hwa, V., Plymate, S. R., Oh, Y. and Rosenfeld, R. G. (2000) Characterization of insulin-like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithelial cells: potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology 141, 4072-4080.   DOI
15 Maki, R. G. (2010) Small is beautiful: insulin-like growth factors and their role in growth, development, and cancer. J. Clin. Oncol. 28, 4985-4995.   DOI   ScienceOn
16 Wang, S., Moerman, E. J., Jones, R. A., Thweatt, R. and Goldstein, S. (1996) Characterization of IGFBP-3, PAI-1 and SPARC mRNA expression in senescent fibroblasts. Mech. Ageing. Dev. 92, 121-132.   DOI   ScienceOn
17 Liu, D. and Hornsby, P. J. (2007) Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Res. 67, 3117-3126.   DOI   ScienceOn
18 Tsai, K. K., Chuang, E. Y., Little, J. B. and Yuan, Z. M. (2005) Cellular mechanisms for low-dose ionizing radiation- induced perturbation of the breast tissue microenvironment. Cancer Res. 65, 6734-6744.   DOI   ScienceOn
19 Dass, K., Ahmad, A., Azmi, A. S., Sarkar, S. H. and Sarkar, F. H. (2008) Evolving role of uPA/uPAR system in human cancers. Cancer Treat. Rev. 34, 122-136.   DOI   ScienceOn
20 Hornebeck, W. and Maquart, F. X. (2003) Proteolyzed matrix as a template for the regulation of tumor progression. Biomed. Pharmacother. 57, 223-230.   DOI   ScienceOn
21 Schadendorf, D., Moller, A., Algermissen, B., Worm, M., Sticherling, M. and Czarnetzki, B. M. (1993) IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. J. Immunol. 151, 2667-2675.
22 Norgauer, J., Metzner, B. and Schraufstatter, I. (1996) Expression and growth-promoting function of the IL-8 receptor beta in human melanoma cells. J. Immunol. 156, 1132-1137.
23 Ksiazek, K., Jorres, A. and Witowski, J. (2008) Senescence induces a proangiogenic switch in human peritoneal mesothelial cells. Rejuvenation. Res. 11, 681-683.   DOI   ScienceOn
24 Chaturvedi, S. and Hass, R. (2011) Extracellular signals in young and aging breast epithelial cells and possible connections to age-associated breast cancer development. Mech. Ageing. Dev. 132, 213-219.   DOI   ScienceOn
25 Studebaker, A. W., Storci, G., Werbeck, J. L., Sansone, P., Sasser, A. K., Tavolari, S., Huang, T., Chan, M. W., Marini, F. C., Rosol, T. J., Bonafe, M. and Hall, B. M. (2008) Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 68, 9087-9095.   DOI   ScienceOn
26 Laberge, R. M., Awad, P., Campisi, J. and Desprez, P. Y. (2012) Epithelial-mesenchymal transition induced by senescent fibroblasts. Cancer Microenviron. 5, 39-44.   DOI   ScienceOn
27 Smit, M. A. and Peeper, D. S. (2010) Epithelial-mesenchymal transition and senescence: two cancer-related processes are crossing paths. Aging (Albany NY) 2, 735-741.
28 Sica, A., Schioppa, T., Mantovani, A. and Allavena, P. (2006) Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur. J. Cancer 42, 717-727.   DOI   ScienceOn
29 Han, N. K., Kim, B. C., Lee, H. C., Lee, Y. J., Park, M. J., Chi, S. G., Ko, Y. G. and Lee, J. S. (2012) Secretome analysis of ionizing radiation-induced senescent cancer cells reveals that secreted RKIP plays a critical role in neighboring cell migration. Proteomics 12, 2822-2832.   DOI   ScienceOn
30 Nardella, C., Clohessy, J. G., Alimonti, A. and Pandolfi, P. P. (2011) Pro-senescence therapy for cancer treatment. Nat. Rev. Cancer 11, 503-511.   DOI   ScienceOn
31 Acosta, J. C. and Gil, J. (2012) Senescence: a new weapon for cancer therapy. Trends. Cell Biol. 22, 211-219.   DOI   ScienceOn
32 Ewald, J. A., Desotelle, J. A., Wilding, G. and Jarrard, D. F. (2010) Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 102, 1536-1546.   DOI   ScienceOn
33 Schwarze, S. R., Fu, V. X., Desotelle, J. A., Kenowski, M. L. and Jarrard, D. F. (2005) The identification of senescence- specific genes during the induction of senescence in prostate cancer cells. Neoplasia 7, 816-823.   DOI   ScienceOn
34 Asai, A., Oshima, Y., Yamamoto, Y., Uochi, T. A., Kusaka, H., Akinaga, S., Yamashita, Y., Pongracz, K., Pruzan, R., Wunder, E., Piatyszek, M., Li, S., Chin, A. C., Harley, C. B. and Gryaznov, S. (2003) A novel telomerase template antagonist (GRN163) as a potential anticancer agent. Cancer Res. 63, 3931-3939.
35 Rebbaa, A., Zheng, X., Chu, F. and Mirkin, B. L. (2006) The role of histone acetylation versus DNA damage in drug-induced senescence and apoptosis. Cell. Death. Differ. 13, 1960-1967.   DOI   ScienceOn
36 Nardella, C., Clohessy, J. G., Alimonti, A. and Pandolfi, P. P. (2011) Pro-senescence therapy for cancer treatment. Nature. Rev. Cancer 11, 503-511.   DOI   ScienceOn
37 Lee, J. J., Kim, B. C., Park, M. J., Lee, Y. S., Kim, Y. N., Lee, B. L. and Lee, J. S. (2011) PTEN status switches cell fate between premature senescence and apoptosis in glioma exposed to ionizing radiation. Cell Death. Differ. 18, 666-677.   DOI   ScienceOn
38 Ventura, A., Kirsch, D. G., McLaughlin, M. E., Tuveson, D. A., Grimm, J., Lintault, L., Newman, J., Reczek, E. E., Weissleder, R. and Jacks, T. (2007) Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661-665.   DOI   ScienceOn
39 Martinkova, E., Maglott, A., Leger, D. Y., Bonnet, D., Stiborova, M., Takeda, K., Martin, S. and Dontenwill, M. (2010) alpha5beta1 integrin antagonists reduce chemotherapy- induced premature senescence and facilitate apoptosis in human glioblastoma cells. Int. J. Cancer 127, 1240-1248.   DOI   ScienceOn
40 Bykov, V. J., Issaeva, N., Shilov, A., Hultcrantz, M., Pugacheva, E., Chumakov, P., Bergman, J., Wiman, K. G. and Selivanova, G. (2002) Restoration of the tumor suppressor function to mutant p53 by a low-molecular-weight compound. Nat. Med. 8, 282-288.   DOI   ScienceOn
41 Michaloglou, C., Vredeveld, L. C., Soengas, M. S., Denoyelle, C., Kuilman, T., van der Horst, C. M., Majoor, D. M., Shay, J. W., Mooi, W. J. and Peeper, D. S. (2005) BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720-724.   DOI   ScienceOn
42 Alimonti, A., Nardella, C., Chen, Z., Clohessy, J. G., Carracedo, A., Trotman, L. C., Cheng, K., Varmeh, S., Kozma, S. C., Thomas, G., Rosivatz, E., Woscholski, R., Cognetti, F., Scher, H. I. and Pandolfi, P. P. (2010) A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J. Clin. Invest. 120, 681-693.   DOI   ScienceOn
43 Delmore, J. E., Issa, G. C., Lemieux, M. E., Rahl, P. B., Shi, J., Jacobs, H. M., Kastritis, E., Gilpatrick, T., Paranal, R. M., Qi, J., Chesi, M., Schinzel, A. C., McKeown, M. R., Heffernan, T. P., Vakoc, C. R., Bergsagel, P. L., Ghobrial, I. M., Richardson, P. G., Young, R. A., Hahn, W. C., Anderson, K. C., Kung, A. L., Bradner, J. E. and Mitsiades, C. S. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904-917.   DOI   ScienceOn
44 Lee, J. J., Lee, J. H., Ko, Y. G., Hong, S. I. and Lee, J. S. (2010) Prevention of premature senescence requires JNK regulation of Bcl-2 and reactive oxygen species. Oncogene 29, 561-575.   DOI   ScienceOn
45 Kim, B. C., Lee, H. C., Lee, J. J., Choi, C. M., Kim, D. K., Lee, J. C., Ko, Y. G. and Lee, J. S. (2012) Wig1 prevents cellular senescence by regulating p21 mRNA decay through control of RISC recruitment. EMBO J. 31, 4289-4303.   DOI
46 Chen, Z., Trotman, L. C., Shaffer, D., Lin, H. K., Dotan, Z. A., Niki, M., Koutcher, J. A., Scher, H. I., Ludwig, T., Gerald, W., Cordon-Cardo, C. and Pandolfi, P. P. (2005) Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725-730.   DOI   ScienceOn
47 Efeyan, A., Ortega-Molina, A., Velasco-Miguel, S., Herranz, D., Vassilev, L. T. and Serrano, M. (2007) Induction of p53-dependent senescence by the MDM2 antagonist nutlin-3a in mouse cells of fibroblast origin. Cancer Res. 67, 7350-7357.   DOI   ScienceOn
48 Campisi, J. (1996) Replicative senescence: an old lives' tale? Cell 84, 497-500.   DOI   ScienceOn
49 Deng, Y. and Chang, S. (2007) Role of telomeres and telomerase in genomic instability, senescence and cancer. Lab. Invest. 87, 1071-1076.   DOI   ScienceOn
50 Toussaint, O., Medrano, E. E. and von Zglinicki, T. (2000) Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp. Gerontol. 35, 927-945.   DOI   ScienceOn
51 Gorgoulis, V. G. and Halazonetis, T. D. (2010) Oncogene-induced senescence: the bright and dark side of the response. Curr. Opin. Cell. Biol. 22, 816-827.   DOI   ScienceOn
52 Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. and Lowe, S. W. (1997) Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593-602.   DOI   ScienceOn
53 Shamma, A., Takegami, Y., Miki, T., Kitajima, S., Noda, M., Obara, T., Okamoto, T. and Takahashi, C. (2009) Rb Regulates DNA damage response and cellular senescence through E2F-dependent suppression of N-ras isoprenylation. Cancer Cell 15, 255-269.   DOI   ScienceOn
54 Suzuki, M. and Boothman, D. A. (2008) Stress-induced premature senescence (SIPS)--influence of SIPS on radiotherapy. J. Radiat. Res. 49, 105-112.   DOI   ScienceOn
55 Young, A. R. and Narita, M. (2009) SASP reflects senescence. EMBO Rep. 10, 228-230.   DOI   ScienceOn
56 Gewirtz, D. A., Holt, S. E. and Elmore, L. W. (2008) Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem. Pharmacol. 76, 947-957.   DOI   ScienceOn
57 Davalos, A. R., Coppe, J. P., Campisi, J. and Desprez, P. Y. (2010) Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis. Rev. 29, 273-283.   DOI   ScienceOn
58 Campisi, J., Andersen, J. K., Kapahi, P. and Melov, S. (2011) Cellular senescence: a link between cancer and age-related degenerative disease? Semin. Cancer Biol. 21, 354-359.
59 Coppe, J. P., Desprez, P. Y., Krtolica, A. and Campisi, J. (2010) The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99-118.   DOI   ScienceOn
60 Campisi, J. (2013) Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75, 685-705.   DOI   ScienceOn
61 de Magalhaes, J. P. (2013) How ageing processes influence cancer. Nat. Rev. Cancer 13, 357-365.   DOI   ScienceOn
62 Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M. and Kroemer, G. (2013) The hallmarks of aging. Cell 153, 1194-1217.   DOI   ScienceOn
63 Oyama, K., Okawa, T., Nakagawa, H., Takaoka, M., Andl, C. D., Kim, S. H., Klein-Szanto, A., Diehl, J. A., Herlyn, M., El-Deiry, W. and Rustgi, A. K. (2007) AKT induces senescence in primary esophageal epithelial cells but is permissive for differentiation as revealed in organotypic culture. Oncogene 26, 2353-2364.   DOI   ScienceOn
64 Dhomen, N., Reis-Filho, J. S., da Rocha Dias, S., Hayward, R., Savage, K., Delmas, V., Larue, L., Pritchard, C. and Marais, R. (2009) Oncogenic Braf induces melanocyte senescene and melanoma in mice. Cancer Cell 15, 294-303.   DOI   ScienceOn
65 Collado, M., Efeyan, A., Guerra, C., Schuhmacher, A. J., Barradas, M., Benguria, A., Zaballos, A., Flores, J. M., Barbacid, M., Beach, D. and Serrano, M. (2005) Tumor biology: senescence in premalignant tumors. Nature 436, 642.   DOI   ScienceOn
66 Kuilman, T., Michaloglou, C., Mooi, W. J. and Peeper, D. S. (2010) The essence of senescence. Genes Dev. 24, 2463-2479.   DOI   ScienceOn
67 Canpo-Trapero, J., Cano-Sanchez, J., Palacios-Sanchez, B., Llamas-Martinez, S., Lo Muzio, L. and Bascones-Martinez, A. (2008) Cellular senescence in oral cancer and precancer and treatment implications: A review. Acta. Oncologica. 47, 1464-1474.   DOI   ScienceOn
68 de Magalhaes, J. P., Curado, J. and Church, G. M. (2009) Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics 25, 875-881.   DOI   ScienceOn
69 Mason, D. X., Jackson, T. J. and Lin, A. W. (2004) Molecular signature of oncogenic ras-induced senescence. Oncogene 23, 9238-9246.   DOI
70 Wennmalm, K., Wahlestedt, C. and Larsson, O. (2005) The expression signature of in vitro senescence resembles mouse but not human aging. Genome. Biol. 6, R109.   DOI
71 Calvanese, V., Lara, E., Kahn, A. and Fraga, M. F. (2009) The role of epigenetics in aging and age-related diseases. Ageing. Res. Rev. 8, 268-276.   DOI   ScienceOn
72 Larsson, L. G. (2011) Oncogene- and tumor suppressor gene-mediated suppression of cellular senescence. Semin. Cancer Biol. 21, 367-376.
73 Ohtani, N. and Hara, E. (2013) Roles and mechanisms of cellular senescence in regulation of tissue homeostasis. Cancer Sci. 104, 525-530.   DOI   ScienceOn
74 Adams, P. D. (2009) Healing and hurting: molecular mechanisms, functions, and pathologies of cellular senescence. Mol. Cell 36, 2-14.   DOI   ScienceOn
75 Chandler, H. and Peters, G. (2013) Stressing the cell cycle in senescence and aging. Curr. Opin. Cell Biol. 25, 765-771.   DOI   ScienceOn
76 Reinhardt, H. C. and Schumacher, B. (2012) The p53 network: cellular and systemic DNA damage responses in aging and cancer. Trends. Genet. 28, 128-136.   DOI   ScienceOn
77 Warfel, N. A. and El-Deiry, W. S. (2013) p21WAF1 and tumourigenesis: 20 years after. Curr. Opin. Oncol. 25, 52-58.   DOI
78 Roninson, I. B. (2002) Oncogenic functions of tumour suppressor p21(Waf1/Cip1/Sdi1): association with cell senescence and tumour-promoting activities of stromal fibroblasts. Cancer Lett. 179, 1-14.   DOI   ScienceOn
79 Carnero, A. and Beach, D. H. (2004) Absence of p21WAF1 cooperates with c-myc in bypassing Ras-induced senescence and enhances oncogenic cooperation. Oncogene 23, 6006-6011.   DOI   ScienceOn
80 Groth, A., Weber, J. D., Willumsen, B. M., Sherr, C. J. and Roussel, M. F. (2000) Oncogenic Ras induces p19ARF and growth arrest in mouse embryo fibroblasts lacking p21Cip1 and p27Kip1 without activating cyclin D-dependent kinases. J. Biol. Chem. 275, 27473-27480.
81 Pantoja, C. and Serrano, M. (1999) Murine fibroblasts lacking p21 undergo senescence and are resistant to transformation by oncogenic Ras. Oncogene 18, 4974-4982.   DOI
82 Dominguez-Brauer, C., Brauer, P. M., Chen, Y. J., Pimkina, J. and Raychaudhuri, P. (2010) Tumor suppression by ARF: gatekeeper and caretaker. Cell Cycle. 9, 86-89.   DOI
83 Kim, W. Y. and Sharpless, N. E. (2006) The regulation of INK4/ARF in cancer and aging. Cell 127, 265-275.   DOI   ScienceOn
84 Rayess, H., Wang, M. B. and Srivatsan, E. S. (2012) Cellular senescence and tumor suppressor gene p16. Int. J. Cancer 130, 1715-1725.   DOI   ScienceOn
85 Kang, T. W., Yevsa, T., Woller, N., Hoenicke, L., Wuestefeld, T., Dauch, D., Hohmeyer, A., Gereke, M., Rudalska, R., Potapova, A., Iken, M., Vucur, M., Weiss, S., Heikenwalder, M., Khan, S., Gil, J., Bruder, D., Manns, M., Schirmacher, P., Tacke, F., Ott, M., Luedde, T., Longerich, T., Kubicka, S. and Zender, L. (2011) Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479, 547-551.   DOI   ScienceOn
86 Benanti, J. A. and Galloway, D. A. (2004) The normal response to RAS: senescence or transformation? Cell Cycle. 3, 715-717.
87 Young, A. P., Schlisio, S., Minamishima, Y. A., Zhang, Q., Li, L., Grisanzio, C., Signoretti, S. and Kaelin, W. G. Jr. (2008) VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400. Nat. Cell Biol. 10, 361-369.   DOI   ScienceOn
88 Efeyan, A., Ortega-Molina, A., Velasco-Miguel, S., Herranz, D., Vassilev, L. T. and Serrano, M. (2007) Induction of p53-dependent senescence by the MDM2 antagonist nutlin-3a in mouse cells of fibroblast origin. Cancer Res. 67, 7350-7357.   DOI   ScienceOn
89 Ben-Porath, I. and Weinberg, R. A. (2005) The signals and pathways activating cellular senescence. Int. J. Biochem. Cell. Biol. 37, 961-976.   DOI   ScienceOn
90 Dimri, G. P., Lee, X., Basile, G., Acosta, M., Scott, G., Roskelley, C., Medrano, E. E., Linskens, M., Rubelj, I., Pereira-Smith, O., Peacocke, M. and Campisi, J. (1995) A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. U. S. A. 92, 9363-9367.   DOI   ScienceOn
91 Byun, H. O., Han, N. K., Lee, H. J., Kim, K. B., Ko, Y. G., Yoon, G., Lee, Y. S., Hong, S. I. and Lee, J. S. (2009) Cathepsin D and eukaryotic translation elongation factor 1 as promising markers of cellular senescence. Cancer Res. 69, 4638-4647.   DOI   ScienceOn
92 Indovina, P., Marcelli, E., Casini, N., Rizzo, V. and Giordano, A. (2013) Emerging roles of RB family: new defense mechanisms against tumor progression. J. Cell Physiol. 228, 525-535.   DOI   ScienceOn
93 Pazolli, E., Luo, X., Brehm, S., Carbery, K., Chung, J. J., Prior, J. L., Doherty, J., Demehri, S., Salavaggione, L., Piwnica-Worms, D. and Stewart, S. A. (2009) Senescent stromal-derived osteopontin promotes preneoplastic cell growth. Cancer Res. 69, 1230-1239.   DOI   ScienceOn
94 Barcellos-Hoff, M. H. and Ravani, S. A. (2000) Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60, 1254-1260.
95 Xue, W., Zender, L., Miethng, C., Dickins R. A., Hernando, E., Krizhanovsky, V., Cordon-Cardo, C. and Lowe, S. M. (2007) Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656-660.   DOI   ScienceOn