Skip to main content
Top
Published in: Tumor Biology 4/2010

01-08-2010 | Review

Radiation-induced cell death mechanisms

Authors: David Eriksson, Torgny Stigbrand

Published in: Tumor Biology | Issue 4/2010

Login to get access

Abstract

The main goal when treating malignancies with radiation therapy is to deprive tumor cells of their reproductive potential. One approach to achieve this is by inducing tumor cell apoptosis. Accumulating evidences suggest that induction of apoptosis alone is insufficient to account for the therapeutic effect of radiotherapy. It has become obvious in the last few years that inhibition of the proliferative capacity of malignant cells following irradiation, especially with solid tumors, can occur via alternative cell death modalities or permanent cell cycle arrests, i.e., senescence. In this review, apoptosis and mitotic catastrophe, the two major cell deaths induced by radiation, are described and dissected in terms of activating mechanisms. Furthermore, treatment-induced senescence and its relevance for the outcome of radiotherapy of cancer will be discussed. The importance of p53 for the induction and execution of these different types of cell deaths is highlighted. The efficiency of radiotherapy and radioimmunotherapy has much to gain by understanding the cell death mechanisms that are induced in tumor cells following irradiation. Strategies to use specific inhibitors that will manipulate key molecules in these pathways in combination with radiation might potentiate therapy and enhance tumor cell kill.
Literature
1.
go back to reference Jonathan EC, Bernhard EJ, McKenna WG. How does radiation kill cells? Curr Opin Chem Biol. 1999;3:77–83.PubMed Jonathan EC, Bernhard EJ, McKenna WG. How does radiation kill cells? Curr Opin Chem Biol. 1999;3:77–83.PubMed
2.
go back to reference Hollstein M, Sidransky D, Vogelstein B, Harris CC. P53 mutations in human cancers. Science. 1991;253:49–53.PubMed Hollstein M, Sidransky D, Vogelstein B, Harris CC. P53 mutations in human cancers. Science. 1991;253:49–53.PubMed
3.
go back to reference Soussi T, Beroud C. Assessing tp53 status in human tumours to evaluate clinical outcome. Nat Rev Cancer. 2001;1:233–40.PubMed Soussi T, Beroud C. Assessing tp53 status in human tumours to evaluate clinical outcome. Nat Rev Cancer. 2001;1:233–40.PubMed
4.
go back to reference Soussi T, Lozano G. P53 mutation heterogeneity in cancer. Biochem Biophys Res Commun. 2005;331:834–42.PubMed Soussi T, Lozano G. P53 mutation heterogeneity in cancer. Biochem Biophys Res Commun. 2005;331:834–42.PubMed
5.
go back to reference Igney FH, Krammer PH. Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer. 2002;2:277–88.PubMed Igney FH, Krammer PH. Death and anti-death: tumour resistance to apoptosis. Nat Rev Cancer. 2002;2:277–88.PubMed
6.
go back to reference Eriksson D, Riklund Ahlstrom K, Johansson L, Stigbrand T. Radiation induced cell deaths. In: Stigbrand T, Carlsson J, Adams G, editors. Targeted radionuclide tumor therapy—biological aspects. New York: Springer; 2008. Eriksson D, Riklund Ahlstrom K, Johansson L, Stigbrand T. Radiation induced cell deaths. In: Stigbrand T, Carlsson J, Adams G, editors. Targeted radionuclide tumor therapy—biological aspects. New York: Springer; 2008.
7.
go back to reference Gewirtz DA, Holt SE, Elmore LW. Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem Pharmacol. 2008;76:947–57.PubMed Gewirtz DA, Holt SE, Elmore LW. Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem Pharmacol. 2008;76:947–57.PubMed
8.
go back to reference Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. P53-dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81:445–58.PubMed Jones KR, Elmore LW, Jackson-Cook C, Demasters G, Povirk LF, Holt SE, et al. P53-dependent accelerated senescence induced by ionizing radiation in breast tumour cells. Int J Radiat Biol. 2005;81:445–58.PubMed
9.
go back to reference Lehmann BD, McCubrey JA, Jefferson HS, Paine MS, Chappell WH, Terrian DM. A dominant role for p53-dependent cellular senescence in radiosensitization of human prostate cancer cells. Cell Cycle. 2007;6:595–605.PubMed Lehmann BD, McCubrey JA, Jefferson HS, Paine MS, Chappell WH, Terrian DM. A dominant role for p53-dependent cellular senescence in radiosensitization of human prostate cancer cells. Cell Cycle. 2007;6:595–605.PubMed
10.
go back to reference Ianzini F, Bertoldo A, Kosmacek EA, Phillips SL, Mackey MA. Lack of p53 function promotes radiation-induced mitotic catastrophe in mouse embryonic fibroblast cells. Cancer Cell Int. 2006;6:11.PubMed Ianzini F, Bertoldo A, Kosmacek EA, Phillips SL, Mackey MA. Lack of p53 function promotes radiation-induced mitotic catastrophe in mouse embryonic fibroblast cells. Cancer Cell Int. 2006;6:11.PubMed
11.
go back to reference Efeyan A, Serrano M. P53: Guardian of the genome and policeman of the oncogenes. Cell Cycle. 2007;6:1006–10.PubMed Efeyan A, Serrano M. P53: Guardian of the genome and policeman of the oncogenes. Cell Cycle. 2007;6:1006–10.PubMed
12.
go back to reference Gottlieb TM, Oren M. P53 in growth control and neoplasia. Biochim Biophys Acta. 1996;1287:77–102.PubMed Gottlieb TM, Oren M. P53 in growth control and neoplasia. Biochim Biophys Acta. 1996;1287:77–102.PubMed
13.
go back to reference Lane DP. Cancer. P53, guardian of the genome. Nature. 1992;358:15–6.PubMed Lane DP. Cancer. P53, guardian of the genome. Nature. 1992;358:15–6.PubMed
14.
go back to reference Marx J. Oncology. Recruiting the cell's own guardian for cancer therapy. Science. 2007;315:1211–3.PubMed Marx J. Oncology. Recruiting the cell's own guardian for cancer therapy. Science. 2007;315:1211–3.PubMed
15.
go back to reference Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and tumor suppression. Nucleic Acids Res. 2007;35:7475–84.PubMed Rodier F, Campisi J, Bhaumik D. Two faces of p53: aging and tumor suppression. Nucleic Acids Res. 2007;35:7475–84.PubMed
16.
go back to reference Teodoro JG, Evans SK, Green MR. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J Mol Med. 2007;85:1175–86.PubMed Teodoro JG, Evans SK, Green MR. Inhibition of tumor angiogenesis by p53: a new role for the guardian of the genome. J Mol Med. 2007;85:1175–86.PubMed
17.
go back to reference Helton ES, Chen X. P53 modulation of the DNA damage response. J Cell Biochem. 2007;100:883–96.PubMed Helton ES, Chen X. P53 modulation of the DNA damage response. J Cell Biochem. 2007;100:883–96.PubMed
18.
go back to reference Aylon Y, Oren M. Living with p53, dying of p53. Cell. 2007;130:597–600.PubMed Aylon Y, Oren M. Living with p53, dying of p53. Cell. 2007;130:597–600.PubMed
19.
go back to reference Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3:117–29.PubMed Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer. 2003;3:117–29.PubMed
20.
go back to reference Harris SL, Levine AJ. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24:2899–908.PubMed Harris SL, Levine AJ. The p53 pathway: positive and negative feedback loops. Oncogene. 2005;24:2899–908.PubMed
21.
go back to reference Laptenko O, Prives C. Transcriptional regulation by p53: one protein, many possibilities. Cell Death Differ. 2006;13:951–61.PubMed Laptenko O, Prives C. Transcriptional regulation by p53: one protein, many possibilities. Cell Death Differ. 2006;13:951–61.PubMed
22.
go back to reference Liebermann DA, Hoffman B, Vesely D. P53 induced growth arrest versus apoptosis and its modulation by survival cytokines. Cell Cycle. 2007;6:166–70.PubMed Liebermann DA, Hoffman B, Vesely D. P53 induced growth arrest versus apoptosis and its modulation by survival cytokines. Cell Cycle. 2007;6:166–70.PubMed
23.
go back to reference Das S, Boswell SA, Aaronson SA, Lee SW. P53 promoter selection: choosing between life and death. Cell Cycle. 2008;7:154–7.PubMed Das S, Boswell SA, Aaronson SA, Lee SW. P53 promoter selection: choosing between life and death. Cell Cycle. 2008;7:154–7.PubMed
24.
go back to reference Das S, Raj L, Zhao B, Kimura Y, Bernstein A, Aaronson SA, et al. Hzf determines cell survival upon genotoxic stress by modulating p53 transactivation. Cell. 2007;130:624–37.PubMed Das S, Raj L, Zhao B, Kimura Y, Bernstein A, Aaronson SA, et al. Hzf determines cell survival upon genotoxic stress by modulating p53 transactivation. Cell. 2007;130:624–37.PubMed
25.
go back to reference Tanaka T, Ohkubo S, Tatsuno I, Prives C. Hcas/cse1l associates with chromatin and regulates expression of select p53 target genes. Cell. 2007;130:638–50.PubMed Tanaka T, Ohkubo S, Tatsuno I, Prives C. Hcas/cse1l associates with chromatin and regulates expression of select p53 target genes. Cell. 2007;130:638–50.PubMed
26.
go back to reference Erenpreisa J, Cragg MS. Cancer: a matter of life cycle? Cell Biol Int. 2007;31:1507–10.PubMed Erenpreisa J, Cragg MS. Cancer: a matter of life cycle? Cell Biol Int. 2007;31:1507–10.PubMed
27.
go back to reference Ganem NJ, Pellman D. Limiting the proliferation of polyploid cells. Cell. 2007;131:437–40.PubMed Ganem NJ, Pellman D. Limiting the proliferation of polyploid cells. Cell. 2007;131:437–40.PubMed
28.
go back to reference Ganem NJ, Storchova Z, Pellman D. Tetraploidy, aneuploidy and cancer. Curr Opin Genet Dev. 2007;17:157–62.PubMed Ganem NJ, Storchova Z, Pellman D. Tetraploidy, aneuploidy and cancer. Curr Opin Genet Dev. 2007;17:157–62.PubMed
29.
go back to reference Storchova Z, Pellman D. From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol. 2004;5:45–54.PubMed Storchova Z, Pellman D. From polyploidy to aneuploidy, genome instability and cancer. Nat Rev Mol Cell Biol. 2004;5:45–54.PubMed
30.
go back to reference Dewey WC, Ling CC, Meyn RE. Radiation-induced apoptosis: relevance to radiotherapy. Int J Radiat Oncol Biol Phys. 1995;33:781–96.PubMed Dewey WC, Ling CC, Meyn RE. Radiation-induced apoptosis: relevance to radiotherapy. Int J Radiat Oncol Biol Phys. 1995;33:781–96.PubMed
31.
go back to reference Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res. 2000;301:133–42.PubMed Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res. 2000;301:133–42.PubMed
32.
go back to reference Radford IR, Murphy TK, Radley JM, Ellis SL. Radiation response of mouse lymphoid and myeloid cell lines. Part ii. Apoptotic death is shown by all lines examined. Int J Radiat Biol. 1994;65:217–27.PubMed Radford IR, Murphy TK, Radley JM, Ellis SL. Radiation response of mouse lymphoid and myeloid cell lines. Part ii. Apoptotic death is shown by all lines examined. Int J Radiat Biol. 1994;65:217–27.PubMed
33.
go back to reference Shinomiya N. New concepts in radiation-induced apoptosis: 'premitotic apoptosis' and 'postmitotic apoptosis'. J Cell Mol Med. 2001;5:240–53.PubMed Shinomiya N. New concepts in radiation-induced apoptosis: 'premitotic apoptosis' and 'postmitotic apoptosis'. J Cell Mol Med. 2001;5:240–53.PubMed
34.
go back to reference Clarke AR, Purdie CA, Harrison DJ, Morris RG, Bird CC, Hooper ML, et al. Thymocyte apoptosis induced by p53-dependent and independent pathways. Nature. 1993;362:849–52.PubMed Clarke AR, Purdie CA, Harrison DJ, Morris RG, Bird CC, Hooper ML, et al. Thymocyte apoptosis induced by p53-dependent and independent pathways. Nature. 1993;362:849–52.PubMed
35.
go back to reference Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. P53 is required for radiation-induced apoptosis in mouse thymocytes. Nature. 1993;362:847–9.PubMed Lowe SW, Schmitt EM, Smith SW, Osborne BA, Jacks T. P53 is required for radiation-induced apoptosis in mouse thymocytes. Nature. 1993;362:847–9.PubMed
36.
go back to reference Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 2004;5:897–907.PubMed Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 2004;5:897–907.PubMed
37.
go back to reference Timmer JC, Salvesen GS. Caspase substrates. Cell Death Differ. 2007;14:66–72.PubMed Timmer JC, Salvesen GS. Caspase substrates. Cell Death Differ. 2007;14:66–72.PubMed
38.
39.
go back to reference Alvarez S, Drane P, Meiller A, Bras M, Deguin-Chambon V, Bouvard V, et al. A comprehensive study of p53 transcriptional activity in thymus and spleen of gamma irradiated mouse: high sensitivity of genes involved in the two main apoptotic pathways. Int J Radiat Biol. 2006;82:761–70.PubMed Alvarez S, Drane P, Meiller A, Bras M, Deguin-Chambon V, Bouvard V, et al. A comprehensive study of p53 transcriptional activity in thymus and spleen of gamma irradiated mouse: high sensitivity of genes involved in the two main apoptotic pathways. Int J Radiat Biol. 2006;82:761–70.PubMed
40.
go back to reference Findley HW, Gu L, Yeager AM, Zhou M. Expression and regulation of bcl-2, bcl-xl, and bax correlate with p53 status and sensitivity to apoptosis in childhood acute lymphoblastic leukemia. Blood. 1997;89:2986–93.PubMed Findley HW, Gu L, Yeager AM, Zhou M. Expression and regulation of bcl-2, bcl-xl, and bax correlate with p53 status and sensitivity to apoptosis in childhood acute lymphoblastic leukemia. Blood. 1997;89:2986–93.PubMed
41.
go back to reference Kobayashi T, Ruan S, Jabbur JR, Consoli U, Clodi K, Shiku H, et al. Differential p53 phosphorylation and activation of apoptosis-promoting genes bax and fas/apo-1 by irradiation and ara-c treatment. Cell Death Differ. 1998;5:584–91.PubMed Kobayashi T, Ruan S, Jabbur JR, Consoli U, Clodi K, Shiku H, et al. Differential p53 phosphorylation and activation of apoptosis-promoting genes bax and fas/apo-1 by irradiation and ara-c treatment. Cell Death Differ. 1998;5:584–91.PubMed
42.
go back to reference Zhan Q, Fan S, Bae I, Guillouf C, Liebermann DA, O'Connor PM, et al. Induction of bax by genotoxic stress in human cells correlates with normal p53 status and apoptosis. Oncogene. 1994;9:3743–51.PubMed Zhan Q, Fan S, Bae I, Guillouf C, Liebermann DA, O'Connor PM, et al. Induction of bax by genotoxic stress in human cells correlates with normal p53 status and apoptosis. Oncogene. 1994;9:3743–51.PubMed
43.
go back to reference Erlacher M, Michalak EM, Kelly PN, Labi V, Niederegger H, Coultas L, et al. Bh3-only proteins puma and bim are rate-limiting for gamma-radiation- and glucocorticoid-induced apoptosis of lymphoid cells in vivo. Blood. 2005;106:4131–8.PubMed Erlacher M, Michalak EM, Kelly PN, Labi V, Niederegger H, Coultas L, et al. Bh3-only proteins puma and bim are rate-limiting for gamma-radiation- and glucocorticoid-induced apoptosis of lymphoid cells in vivo. Blood. 2005;106:4131–8.PubMed
44.
go back to reference Michalak EM, Villunger A, Adams JM, Strasser A. In several cell types tumour suppressor p53 induces apoptosis largely via puma but noxa can contribute. Cell Death Differ. 2008;15:1019–29.PubMed Michalak EM, Villunger A, Adams JM, Strasser A. In several cell types tumour suppressor p53 induces apoptosis largely via puma but noxa can contribute. Cell Death Differ. 2008;15:1019–29.PubMed
45.
go back to reference Jeffers JR, Parganas E, Lee Y, Yang C, Wang J, Brennan J, et al. Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell. 2003;4:321–8.PubMed Jeffers JR, Parganas E, Lee Y, Yang C, Wang J, Brennan J, et al. Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell. 2003;4:321–8.PubMed
46.
go back to reference Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, et al. P53- and drug-induced apoptotic responses mediated by bh3-only proteins puma and noxa. Science. 2003;302:1036–8.PubMed Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, et al. P53- and drug-induced apoptotic responses mediated by bh3-only proteins puma and noxa. Science. 2003;302:1036–8.PubMed
47.
go back to reference Lin Y, Ma W, Benchimol S. Pidd, a new death-domain-containing protein, is induced by p53 and promotes apoptosis. Nat Genet. 2000;26:122–7.PubMed Lin Y, Ma W, Benchimol S. Pidd, a new death-domain-containing protein, is induced by p53 and promotes apoptosis. Nat Genet. 2000;26:122–7.PubMed
48.
go back to reference Fei P, El-Deiry WS. P53 and radiation responses. Oncogene. 2003;22:5774–83.PubMed Fei P, El-Deiry WS. P53 and radiation responses. Oncogene. 2003;22:5774–83.PubMed
49.
go back to reference Haldar S, Negrini M, Monne M, Sabbioni S, Croce CM. Down-regulation of bcl-2 by p53 in breast cancer cells. Cancer Res. 1994;54:2095–7.PubMed Haldar S, Negrini M, Monne M, Sabbioni S, Croce CM. Down-regulation of bcl-2 by p53 in breast cancer cells. Cancer Res. 1994;54:2095–7.PubMed
50.
go back to reference Miyashita T, Harigai M, Hanada M, Reed JC. Identification of a p53-dependent negative response element in the bcl-2 gene. Cancer Res. 1994;54:3131–5.PubMed Miyashita T, Harigai M, Hanada M, Reed JC. Identification of a p53-dependent negative response element in the bcl-2 gene. Cancer Res. 1994;54:3131–5.PubMed
51.
go back to reference Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M. Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J Biol Chem. 2002;277:3247–57.PubMed Hoffman WH, Biade S, Zilfou JT, Chen J, Murphy M. Transcriptional repression of the anti-apoptotic survivin gene by wild type p53. J Biol Chem. 2002;277:3247–57.PubMed
52.
go back to reference Zhou M, Gu L, Li F, Zhu Y, Woods WG, Findley HW. DNA damage induces a novel p53-survivin signaling pathway regulating cell cycle and apoptosis in acute lymphoblastic leukemia cells. J Pharmacol Exp Ther. 2002;303:124–31.PubMed Zhou M, Gu L, Li F, Zhu Y, Woods WG, Findley HW. DNA damage induces a novel p53-survivin signaling pathway regulating cell cycle and apoptosis in acute lymphoblastic leukemia cells. J Pharmacol Exp Ther. 2002;303:124–31.PubMed
53.
go back to reference Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15:2922–33.PubMed Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15:2922–33.PubMed
54.
go back to reference Jiang X, Wang X. Cytochrome c promotes caspase-9 activation by inducing nucleotide binding to apaf-1. J Biol Chem. 2000;275:31199–203.PubMed Jiang X, Wang X. Cytochrome c promotes caspase-9 activation by inducing nucleotide binding to apaf-1. J Biol Chem. 2000;275:31199–203.PubMed
55.
go back to reference Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and datp-dependent formation of apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–89.PubMed Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, et al. Cytochrome c and datp-dependent formation of apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–89.PubMed
56.
go back to reference Rodriguez J, Lazebnik Y. Caspase-9 and apaf-1 form an active holoenzyme. Genes Dev. 1999;13:3179–84.PubMed Rodriguez J, Lazebnik Y. Caspase-9 and apaf-1 form an active holoenzyme. Genes Dev. 1999;13:3179–84.PubMed
57.
go back to reference Embree-Ku M, Venturini D, Boekelheide K. Fas is involved in the p53-dependent apoptotic response to ionizing radiation in mouse testis. Biol Reprod. 2002;66:1456–61.PubMed Embree-Ku M, Venturini D, Boekelheide K. Fas is involved in the p53-dependent apoptotic response to ionizing radiation in mouse testis. Biol Reprod. 2002;66:1456–61.PubMed
58.
go back to reference Sheard MA, Uldrijan S, Vojtesek B. Role of p53 in regulating constitutive and x-radiation-inducible cd95 expression and function in carcinoma cells. Cancer Res. 2003;63:7176–84.PubMed Sheard MA, Uldrijan S, Vojtesek B. Role of p53 in regulating constitutive and x-radiation-inducible cd95 expression and function in carcinoma cells. Cancer Res. 2003;63:7176–84.PubMed
59.
go back to reference Sheard MA, Vojtesek B, Janakova L, Kovarik J, Zaloudik J. Up-regulation of fas (cd95) in human p53wild-type cancer cells treated with ionizing radiation. Int J Cancer. 1997;73:757–62.PubMed Sheard MA, Vojtesek B, Janakova L, Kovarik J, Zaloudik J. Up-regulation of fas (cd95) in human p53wild-type cancer cells treated with ionizing radiation. Int J Cancer. 1997;73:757–62.PubMed
60.
go back to reference Burns TF, Bernhard EJ, El-Deiry WS. Tissue specific expression of p53 target genes suggests a key role for killer/dr5 in p53-dependent apoptosis in vivo. Oncogene. 2001;20:4601–12.PubMed Burns TF, Bernhard EJ, El-Deiry WS. Tissue specific expression of p53 target genes suggests a key role for killer/dr5 in p53-dependent apoptosis in vivo. Oncogene. 2001;20:4601–12.PubMed
61.
go back to reference Sheikh MS, Burns TF, Huang Y, Wu GS, Amundson S, Brooks KS, et al. P53-dependent and -independent regulation of the death receptor killer/dr5 gene expression in response to genotoxic stress and tumor necrosis factor alpha. Cancer Res. 1998;58:1593–8.PubMed Sheikh MS, Burns TF, Huang Y, Wu GS, Amundson S, Brooks KS, et al. P53-dependent and -independent regulation of the death receptor killer/dr5 gene expression in response to genotoxic stress and tumor necrosis factor alpha. Cancer Res. 1998;58:1593–8.PubMed
62.
go back to reference Wu GS, Burns TF, McDonald 3rd ER, Jiang W, Meng R, Krantz ID, et al. Killer/dr5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet. 1997;17:141–3.PubMed Wu GS, Burns TF, McDonald 3rd ER, Jiang W, Meng R, Krantz ID, et al. Killer/dr5 is a DNA damage-inducible p53-regulated death receptor gene. Nat Genet. 1997;17:141–3.PubMed
63.
go back to reference Kastan M. On the trail from p53 to apoptosis? Nat Genet. 1997;17:130–1.PubMed Kastan M. On the trail from p53 to apoptosis? Nat Genet. 1997;17:130–1.PubMed
64.
go back to reference Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, et al. Two cd95 (apo-1/fas) signaling pathways. EMBO J. 1998;17:1675–87.PubMed Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ, et al. Two cd95 (apo-1/fas) signaling pathways. EMBO J. 1998;17:1675–87.PubMed
65.
go back to reference Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2007;9:231–41. Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2007;9:231–41.
66.
go back to reference Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94:481–90.PubMed Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell. 1998;94:481–90.PubMed
67.
go back to reference Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L, et al. Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 2007;14:1237–43.PubMed Galluzzi L, Maiuri MC, Vitale I, Zischka H, Castedo M, Zitvogel L, et al. Cell death modalities: Classification and pathophysiological implications. Cell Death Differ. 2007;14:1237–43.PubMed
68.
go back to reference Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat. 2001;4:303–13.PubMed Roninson IB, Broude EV, Chang BD. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist Updat. 2001;4:303–13.PubMed
69.
go back to reference Bhattathiri NV, Bharathykkutty C, Prathapan R, Chirayathmanjiyil DA, Nair KM. Prediction of radiosensitivity of oral cancers by serial cytological assay of nuclear changes. Radiother Oncol. 1998;49:61–5.PubMed Bhattathiri NV, Bharathykkutty C, Prathapan R, Chirayathmanjiyil DA, Nair KM. Prediction of radiosensitivity of oral cancers by serial cytological assay of nuclear changes. Radiother Oncol. 1998;49:61–5.PubMed
70.
go back to reference Bhattathiri NV, Bindu L, Remani P, Chandralekha B, Nair KM. Radiation-induced acute immediate nuclear abnormalities in oral cancer cells: serial cytologic evaluation. Acta Cytol. 1998;42:1084–90.PubMed Bhattathiri NV, Bindu L, Remani P, Chandralekha B, Nair KM. Radiation-induced acute immediate nuclear abnormalities in oral cancer cells: serial cytologic evaluation. Acta Cytol. 1998;42:1084–90.PubMed
71.
go back to reference Eriksson D, Joniani HM, Sheikholvaezin A, Lofroth PO, Johansson L. Combined low dose radio- and radioimmunotherapy of experimental hela hep 2 tumours. Eur J Nucl Med Mol Imaging. 2003;30:895–906.PubMed Eriksson D, Joniani HM, Sheikholvaezin A, Lofroth PO, Johansson L. Combined low dose radio- and radioimmunotherapy of experimental hela hep 2 tumours. Eur J Nucl Med Mol Imaging. 2003;30:895–906.PubMed
72.
go back to reference Eriksson D, Lofroth PO, Johansson L, Riklund KA, Stigbrand T. Cell cycle disturbances and mitotic catastrophes in hela hep2 cells following 2.5 to 10 gy of ionizing radiation. Clin Cancer Res. 2007;13:5501s–8.PubMed Eriksson D, Lofroth PO, Johansson L, Riklund KA, Stigbrand T. Cell cycle disturbances and mitotic catastrophes in hela hep2 cells following 2.5 to 10 gy of ionizing radiation. Clin Cancer Res. 2007;13:5501s–8.PubMed
73.
go back to reference Somosy Z. Radiation response of cell organelles. Micron. 2000;31:165–81.PubMed Somosy Z. Radiation response of cell organelles. Micron. 2000;31:165–81.PubMed
74.
go back to reference Castedo M, Kroemer G. [mitotic catastrophe: a special case of apoptosis]. J Soc Biol. 2004;198:97–103.PubMed Castedo M, Kroemer G. [mitotic catastrophe: a special case of apoptosis]. J Soc Biol. 2004;198:97–103.PubMed
75.
go back to reference Erenpreisa J, Kalejs M, Ianzini F, Kosmacek EA, Mackey MA, Emzinsh D, et al. Segregation of genomes in polyploid tumour cells following mitotic catastrophe. Cell Biol Int. 2005;29:1005–11.PubMed Erenpreisa J, Kalejs M, Ianzini F, Kosmacek EA, Mackey MA, Emzinsh D, et al. Segregation of genomes in polyploid tumour cells following mitotic catastrophe. Cell Biol Int. 2005;29:1005–11.PubMed
76.
go back to reference Bourke E, Dodson H, Merdes A, Cuffe L, Zachos G, Walker M, et al. DNA damage induces chk1-dependent centrosome amplification. EMBO Rep. 2007;8:603–9.PubMed Bourke E, Dodson H, Merdes A, Cuffe L, Zachos G, Walker M, et al. DNA damage induces chk1-dependent centrosome amplification. EMBO Rep. 2007;8:603–9.PubMed
77.
go back to reference Dodson H, Wheatley SP, Morrison CG. Involvement of centrosome amplification in radiation-induced mitotic catastrophe. Cell Cycle. 2007;6:364–70.PubMed Dodson H, Wheatley SP, Morrison CG. Involvement of centrosome amplification in radiation-induced mitotic catastrophe. Cell Cycle. 2007;6:364–70.PubMed
78.
go back to reference Kawamura K, Fujikawa-Yamamoto K, Ozaki M, Iwabuchi K, Nakashima H, Domiki C, et al. Centrosome hyperamplification and chromosomal damage after exposure to radiation. Oncology. 2004;67:460–70.PubMed Kawamura K, Fujikawa-Yamamoto K, Ozaki M, Iwabuchi K, Nakashima H, Domiki C, et al. Centrosome hyperamplification and chromosomal damage after exposure to radiation. Oncology. 2004;67:460–70.PubMed
79.
go back to reference Kawamura K, Morita N, Domiki C, Fujikawa-Yamamoto K, Hashimoto M, Iwabuchi K, et al. Induction of centrosome amplification in p53 sirna-treated human fibroblast cells by radiation exposure. Cancer Sci. 2006;97:252–8.PubMed Kawamura K, Morita N, Domiki C, Fujikawa-Yamamoto K, Hashimoto M, Iwabuchi K, et al. Induction of centrosome amplification in p53 sirna-treated human fibroblast cells by radiation exposure. Cancer Sci. 2006;97:252–8.PubMed
80.
go back to reference Eriksson D, Lofroth PO, Johansson L, Riklund K, Stigbrand T. Apoptotic signalling in hela hep2 cells following 5 gy of cobalt-60 gamma radiation. Anticancer Res. 2009;29:4361–6.PubMed Eriksson D, Lofroth PO, Johansson L, Riklund K, Stigbrand T. Apoptotic signalling in hela hep2 cells following 5 gy of cobalt-60 gamma radiation. Anticancer Res. 2009;29:4361–6.PubMed
81.
go back to reference Loffler H, Lukas J, Bartek J, Kramer A. Structure meets function–centrosomes, genome maintenance and the DNA damage response. Exp Cell Res. 2006;312:2633–40.PubMed Loffler H, Lukas J, Bartek J, Kramer A. Structure meets function–centrosomes, genome maintenance and the DNA damage response. Exp Cell Res. 2006;312:2633–40.PubMed
82.
go back to reference Eriksson D, Blomberg J, Lindgren T, Lofroth PO, Johansson L, Riklund K, et al. Iodine-131 induces mitotic catastrophes and activates apoptotic pathways in hela hep2 cells. Cancer Biother Radiopharm. 2008;23:541–9.PubMed Eriksson D, Blomberg J, Lindgren T, Lofroth PO, Johansson L, Riklund K, et al. Iodine-131 induces mitotic catastrophes and activates apoptotic pathways in hela hep2 cells. Cancer Biother Radiopharm. 2008;23:541–9.PubMed
83.
go back to reference Wang Y, Ji P, Liu J, Broaddus RR, Xue F, Zhang W. Centrosome-associated regulators of the g(2)/m checkpoint as targets for cancer therapy. Mol Cancer. 2009;8:8.PubMed Wang Y, Ji P, Liu J, Broaddus RR, Xue F, Zhang W. Centrosome-associated regulators of the g(2)/m checkpoint as targets for cancer therapy. Mol Cancer. 2009;8:8.PubMed
84.
go back to reference Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37.PubMed Castedo M, Perfettini JL, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37.PubMed
85.
go back to reference Hanashiro K, Kanai M, Geng Y, Sicinski P, Fukasawa K. Roles of cyclins a and e in induction of centrosome amplification in p53-compromised cells. Oncogene. 2008;27:5288–302.PubMed Hanashiro K, Kanai M, Geng Y, Sicinski P, Fukasawa K. Roles of cyclins a and e in induction of centrosome amplification in p53-compromised cells. Oncogene. 2008;27:5288–302.PubMed
86.
go back to reference Fukasawa K. Oncogenes and tumour suppressors take on centrosomes. Nat Rev Cancer. 2007;7:911–24.PubMed Fukasawa K. Oncogenes and tumour suppressors take on centrosomes. Nat Rev Cancer. 2007;7:911–24.PubMed
87.
go back to reference Fukasawa K. P53, cyclin-dependent kinase and abnormal amplification of centrosomes. Biochim Biophys Acta. 2008;1786:15–23.PubMed Fukasawa K. P53, cyclin-dependent kinase and abnormal amplification of centrosomes. Biochim Biophys Acta. 2008;1786:15–23.PubMed
88.
go back to reference Castedo M, Perfettini JL, Roumier T, Valent A, Raslova H, Yakushijin K, et al. Mitotic catastrophe constitutes a special case of apoptosis whose suppression entails aneuploidy. Oncogene. 2004;23:4362–70.PubMed Castedo M, Perfettini JL, Roumier T, Valent A, Raslova H, Yakushijin K, et al. Mitotic catastrophe constitutes a special case of apoptosis whose suppression entails aneuploidy. Oncogene. 2004;23:4362–70.PubMed
89.
go back to reference Abend M. Reasons to reconsider the significance of apoptosis for cancer therapy. Int J Radiat Biol. 2003;79:927–41.PubMed Abend M. Reasons to reconsider the significance of apoptosis for cancer therapy. Int J Radiat Biol. 2003;79:927–41.PubMed
90.
go back to reference Ruth AC, Roninson IB. Effects of the multidrug transporter p-glycoprotein on cellular responses to ionizing radiation. Cancer Res. 2000;60:2576–8.PubMed Ruth AC, Roninson IB. Effects of the multidrug transporter p-glycoprotein on cellular responses to ionizing radiation. Cancer Res. 2000;60:2576–8.PubMed
91.
go back to reference Jallepalli PV, Lengauer C. Chromosome segregation and cancer: cutting through the mystery. Nat Rev Cancer. 2001;1:109–17.PubMed Jallepalli PV, Lengauer C. Chromosome segregation and cancer: cutting through the mystery. Nat Rev Cancer. 2001;1:109–17.PubMed
92.
go back to reference Norbury CJ, Zhivotovsky B. DNA damage-induced apoptosis. Oncogene. 2004;23:2797–808.PubMed Norbury CJ, Zhivotovsky B. DNA damage-induced apoptosis. Oncogene. 2004;23:2797–808.PubMed
93.
go back to reference Zhivotovsky B, Orrenius S. Caspase-2 function in response to DNA damage. Biochem Biophys Res Commun. 2005;331:859–67.PubMed Zhivotovsky B, Orrenius S. Caspase-2 function in response to DNA damage. Biochem Biophys Res Commun. 2005;331:859–67.PubMed
94.
go back to reference Weaver BA, Cleveland DW. Decoding the links between mitosis, cancer, and chemotherapy: the mitotic checkpoint, adaptation, and cell death. Cancer Cell. 2005;8:7–12.PubMed Weaver BA, Cleveland DW. Decoding the links between mitosis, cancer, and chemotherapy: the mitotic checkpoint, adaptation, and cell death. Cancer Cell. 2005;8:7–12.PubMed
95.
go back to reference Yamada HY, Gorbsky GJ. Spindle checkpoint function and cellular sensitivity to antimitotic drugs. Mol Cancer Ther. 2006;5:2963–9.PubMed Yamada HY, Gorbsky GJ. Spindle checkpoint function and cellular sensitivity to antimitotic drugs. Mol Cancer Ther. 2006;5:2963–9.PubMed
96.
go back to reference Rieder CL, Maiato H. Stuck in division or passing through: What happens when cells cannot satisfy the spindle assembly checkpoint. Dev Cell. 2004;7:637–51.PubMed Rieder CL, Maiato H. Stuck in division or passing through: What happens when cells cannot satisfy the spindle assembly checkpoint. Dev Cell. 2004;7:637–51.PubMed
97.
go back to reference Castedo M, Coquelle A, Vivet S, Vitale I, Kauffmann A, Dessen P, et al. Apoptosis regulation in tetraploid cancer cells. EMBO J. 2006;25:2584–95.PubMed Castedo M, Coquelle A, Vivet S, Vitale I, Kauffmann A, Dessen P, et al. Apoptosis regulation in tetraploid cancer cells. EMBO J. 2006;25:2584–95.PubMed
98.
go back to reference Uetake Y, Sluder G. Cell cycle progression after cleavage failure: mammalian somatic cells do not possess a "Tetraploidy checkpoint". J Cell Biol. 2004;165:609–15.PubMed Uetake Y, Sluder G. Cell cycle progression after cleavage failure: mammalian somatic cells do not possess a "Tetraploidy checkpoint". J Cell Biol. 2004;165:609–15.PubMed
99.
go back to reference Blagosklonny MV, Demidenko ZN, Giovino M, Szynal C, Donskoy E, Herrmann RA, et al. Cytostatic activity of paclitaxel in coronary artery smooth muscle cells is mediated through transient mitotic arrest followed by permanent post-mitotic arrest: comparison with cancer cells. Cell Cycle. 2006;5:1574–9.PubMed Blagosklonny MV, Demidenko ZN, Giovino M, Szynal C, Donskoy E, Herrmann RA, et al. Cytostatic activity of paclitaxel in coronary artery smooth muscle cells is mediated through transient mitotic arrest followed by permanent post-mitotic arrest: comparison with cancer cells. Cell Cycle. 2006;5:1574–9.PubMed
100.
go back to reference Klein LE, Freeze BS, Smith 3rd AB, Horwitz SB. The microtubule stabilizing agent discodermolide is a potent inducer of accelerated cell senescence. Cell Cycle. 2005;4:501–7.PubMed Klein LE, Freeze BS, Smith 3rd AB, Horwitz SB. The microtubule stabilizing agent discodermolide is a potent inducer of accelerated cell senescence. Cell Cycle. 2005;4:501–7.PubMed
101.
go back to reference Casenghi M, Mangiacasale R, Tuynder M, Caillet-Fauquet P, Elhajouji A, Lavia P, et al. P53-independent apoptosis and p53-dependent block of DNA rereplication following mitotic spindle inhibition in human cells. Exp Cell Res. 1999;250:339–50.PubMed Casenghi M, Mangiacasale R, Tuynder M, Caillet-Fauquet P, Elhajouji A, Lavia P, et al. P53-independent apoptosis and p53-dependent block of DNA rereplication following mitotic spindle inhibition in human cells. Exp Cell Res. 1999;250:339–50.PubMed
102.
go back to reference Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, et al. A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 1999;59:3761–7.PubMed Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, et al. A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 1999;59:3761–7.PubMed
103.
go back to reference Chang BD, Swift ME, Shen M, Fang J, Broude EV, Roninson IB. Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proc Natl Acad Sci U S A. 2002;99:389–94.PubMed Chang BD, Swift ME, Shen M, Fang J, Broude EV, Roninson IB. Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proc Natl Acad Sci U S A. 2002;99:389–94.PubMed
104.
go back to reference Elmore LW, Rehder CW, Di X, McChesney PA, Jackson-Cook CK, Gewirtz DA, et al. Adriamycin-induced senescence in breast tumor cells involves functional p53 and telomere dysfunction. J Biol Chem. 2002;277:35509–15.PubMed Elmore LW, Rehder CW, Di X, McChesney PA, Jackson-Cook CK, Gewirtz DA, et al. Adriamycin-induced senescence in breast tumor cells involves functional p53 and telomere dysfunction. J Biol Chem. 2002;277:35509–15.PubMed
105.
go back to reference Han Z, Wei W, Dunaway S, Darnowski JW, Calabresi P, Sedivy J, et al. Role of p21 in apoptosis and senescence of human colon cancer cells treated with camptothecin. J Biol Chem. 2002;277:17154–60.PubMed Han Z, Wei W, Dunaway S, Darnowski JW, Calabresi P, Sedivy J, et al. Role of p21 in apoptosis and senescence of human colon cancer cells treated with camptothecin. J Biol Chem. 2002;277:17154–60.PubMed
106.
go back to reference Haq R, Brenton JD, Takahashi M, Finan D, Finkielsztein A, Damaraju S, et al. Constitutive p38hog mitogen-activated protein kinase activation induces permanent cell cycle arrest and senescence. Cancer Res. 2002;62:5076–82.PubMed Haq R, Brenton JD, Takahashi M, Finan D, Finkielsztein A, Damaraju S, et al. Constitutive p38hog mitogen-activated protein kinase activation induces permanent cell cycle arrest and senescence. Cancer Res. 2002;62:5076–82.PubMed
107.
go back to reference Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type tp53. Radiat Res. 2005;163:53–62.PubMed Mirzayans R, Scott A, Cameron M, Murray D. Induction of accelerated senescence by gamma radiation in human solid tumor-derived cell lines expressing wild-type tp53. Radiat Res. 2005;163:53–62.PubMed
108.
go back to reference Quick QA, Gewirtz DA. An accelerated senescence response to radiation in wild-type p53 glioblastoma multiforme cells. J Neurosurg. 2006;105:111–8.PubMed Quick QA, Gewirtz DA. An accelerated senescence response to radiation in wild-type p53 glioblastoma multiforme cells. J Neurosurg. 2006;105:111–8.PubMed
109.
go back to reference Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005;65:2795–803.PubMed Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005;65:2795–803.PubMed
110.
go back to reference Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63:2705–15.PubMed Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63:2705–15.PubMed
111.
go back to reference Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775:5–20.PubMed Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775:5–20.PubMed
112.
go back to reference Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, et al. A senescence program controlled by p53 and p16ink4a contributes to the outcome of cancer therapy. Cell. 2002;109:335–46.PubMed Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, et al. A senescence program controlled by p53 and p16ink4a contributes to the outcome of cancer therapy. Cell. 2002;109:335–46.PubMed
113.
go back to reference Schwarze SR, Fu VX, Desotelle JA, Kenowski ML, Jarrard DF. The identification of senescence-specific genes during the induction of senescence in prostate cancer cells. Neoplasia. 2005;7:816–23.PubMed Schwarze SR, Fu VX, Desotelle JA, Kenowski ML, Jarrard DF. The identification of senescence-specific genes during the induction of senescence in prostate cancer cells. Neoplasia. 2005;7:816–23.PubMed
114.
go back to reference te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP. DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 2002;62:1876–83. te Poele RH, Okorokov AL, Jardine L, Cummings J, Joel SP. DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 2002;62:1876–83.
115.
go back to reference Bataini JP, Belloir C, Mazabraud A, Pilleron JP, Cartigny A, Jaulerry C, et al. Desmoid tumors in adults: The role of radiotherapy in their management. Am J Surg. 1988;155:754–60.PubMed Bataini JP, Belloir C, Mazabraud A, Pilleron JP, Cartigny A, Jaulerry C, et al. Desmoid tumors in adults: The role of radiotherapy in their management. Am J Surg. 1988;155:754–60.PubMed
116.
go back to reference Cox JD, Kline RW. Do prostatic biopsies 12 months or more after external irradiation for adenocarcinoma, stage iii, predict long-term survival? Int J Radiat Oncol Biol Phys. 1983;9:299–303.PubMed Cox JD, Kline RW. Do prostatic biopsies 12 months or more after external irradiation for adenocarcinoma, stage iii, predict long-term survival? Int J Radiat Oncol Biol Phys. 1983;9:299–303.PubMed
117.
go back to reference Shay JW, Roninson IB. Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004;23:2919–33.PubMed Shay JW, Roninson IB. Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004;23:2919–33.PubMed
118.
go back to reference Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: The senescence connection. Nat Rev Cancer. 2008;8:450–8.PubMed Deng Y, Chan SS, Chang S. Telomere dysfunction and tumour suppression: The senescence connection. Nat Rev Cancer. 2008;8:450–8.PubMed
119.
go back to reference Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961;25:585–621.
120.
go back to reference Stein GH, Dulic V. Origins of g1 arrest in senescent human fibroblasts. Bioessays. 1995;17:537–43.PubMed Stein GH, Dulic V. Origins of g1 arrest in senescent human fibroblasts. Bioessays. 1995;17:537–43.PubMed
121.
go back to reference Collado M, Serrano M. Senescence in tumours: evidence from mice and humans. Nat Rev Cancer. 2010;10:51–7.PubMed Collado M, Serrano M. Senescence in tumours: evidence from mice and humans. Nat Rev Cancer. 2010;10:51–7.PubMed
122.
go back to reference Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11:973–9.PubMed Rodier F, Coppe JP, Patil CK, Hoeijmakers WA, Munoz DP, Raza SR, et al. Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat Cell Biol. 2009;11:973–9.PubMed
123.
go back to reference Suzuki K, Mori I, Nakayama Y, Miyakoda M, Kodama S, Watanabe M. Radiation-induced senescence-like growth arrest requires tp53 function but not telomere shortening. Radiat Res. 2001;155:248–53.PubMed Suzuki K, Mori I, Nakayama Y, Miyakoda M, Kodama S, Watanabe M. Radiation-induced senescence-like growth arrest requires tp53 function but not telomere shortening. Radiat Res. 2001;155:248–53.PubMed
124.
go back to reference Dai CY, Enders GH. P16 ink4a can initiate an autonomous senescence program. Oncogene. 2000;19:1613–22.PubMed Dai CY, Enders GH. P16 ink4a can initiate an autonomous senescence program. Oncogene. 2000;19:1613–22.PubMed
125.
go back to reference Steiner MS, Wang Y, Zhang Y, Zhang X, Lu Y. P16/mts1/ink4a suppresses prostate cancer by both prb dependent and independent pathways. Oncogene. 2000;19:1297–306.PubMed Steiner MS, Wang Y, Zhang Y, Zhang X, Lu Y. P16/mts1/ink4a suppresses prostate cancer by both prb dependent and independent pathways. Oncogene. 2000;19:1297–306.PubMed
126.
go back to reference Sugrue MM, Shin DY, Lee SW, Aaronson SA. Wild-type p53 triggers a rapid senescence program in human tumor cells lacking functional p53. Proc Natl Acad Sci U S A. 1997;94:9648–53.PubMed Sugrue MM, Shin DY, Lee SW, Aaronson SA. Wild-type p53 triggers a rapid senescence program in human tumor cells lacking functional p53. Proc Natl Acad Sci U S A. 1997;94:9648–53.PubMed
127.
go back to reference Zhang H. Molecular signaling and genetic pathways of senescence: its role in tumorigenesis and aging. J Cell Physiol. 2007;210:567–74.PubMed Zhang H. Molecular signaling and genetic pathways of senescence: its role in tumorigenesis and aging. J Cell Physiol. 2007;210:567–74.PubMed
128.
go back to reference Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.PubMed Coppe JP, Desprez PY, Krtolica A, Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118.PubMed
129.
go back to reference Novakova Z, Hubackova S, Kosar M, Janderova-Rossmeislova L, Dobrovolna J, Vasicova P, et al. Cytokine expression and signaling in drug-induced cellular senescence. Oncogene. 2010;29:273–84.PubMed Novakova Z, Hubackova S, Kosar M, Janderova-Rossmeislova L, Dobrovolna J, Vasicova P, et al. Cytokine expression and signaling in drug-induced cellular senescence. Oncogene. 2010;29:273–84.PubMed
130.
go back to reference Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.PubMed Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.PubMed
131.
go back to reference Bassett EA, Wang W, Rastinejad F, El-Deiry WS. Structural and functional basis for therapeutic modulation of p53 signaling. Clin Cancer Res. 2008;14:6376–86.PubMed Bassett EA, Wang W, Rastinejad F, El-Deiry WS. Structural and functional basis for therapeutic modulation of p53 signaling. Clin Cancer Res. 2008;14:6376–86.PubMed
132.
go back to reference Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer. 2009;9:862–73.PubMed Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer. 2009;9:862–73.PubMed
133.
go back to reference Wang W, El-Deiry WS. Restoration of p53 to limit tumor growth. Curr Opin Oncol. 2008;20:90–6.PubMed Wang W, El-Deiry WS. Restoration of p53 to limit tumor growth. Curr Opin Oncol. 2008;20:90–6.PubMed
134.
go back to reference Wiman KG. Restoration of wild-type p53 function in human tumors: Strategies for efficient cancer therapy. Adv Cancer Res. 2007;97:321–38.PubMed Wiman KG. Restoration of wild-type p53 function in human tumors: Strategies for efficient cancer therapy. Adv Cancer Res. 2007;97:321–38.PubMed
135.
go back to reference Kawabe T. G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther. 2004;3:513–9.PubMed Kawabe T. G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther. 2004;3:513–9.PubMed
136.
go back to reference Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8:547–66.PubMed Lapenna S, Giordano A. Cell cycle kinases as therapeutic targets for cancer. Nat Rev Drug Discov. 2009;8:547–66.PubMed
137.
go back to reference Gautschi O, Heighway J, Mack PC, Purnell PR, Lara Jr PN, Gandara DR. Aurora kinases as anticancer drug targets. Clin Cancer Res. 2008;14:1639–48.PubMed Gautschi O, Heighway J, Mack PC, Purnell PR, Lara Jr PN, Gandara DR. Aurora kinases as anticancer drug targets. Clin Cancer Res. 2008;14:1639–48.PubMed
138.
go back to reference Kitzen JJ, de Jonge MJ, Verweij J. Aurora kinase inhibitors. Crit Rev Oncol Hematol. 2010;73:99–110.PubMed Kitzen JJ, de Jonge MJ, Verweij J. Aurora kinase inhibitors. Crit Rev Oncol Hematol. 2010;73:99–110.PubMed
139.
go back to reference Mita AC, Mita MM, Nawrocki ST, Giles FJ. Survivin: Key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res. 2008;14:5000–5.PubMed Mita AC, Mita MM, Nawrocki ST, Giles FJ. Survivin: Key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res. 2008;14:5000–5.PubMed
140.
go back to reference Kwon M, Godinho SA, Chandhok NS, Ganem NJ, Azioune A, Thery M, et al. Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008;22:2189–203.PubMed Kwon M, Godinho SA, Chandhok NS, Ganem NJ, Azioune A, Thery M, et al. Mechanisms to suppress multipolar divisions in cancer cells with extra centrosomes. Genes Dev. 2008;22:2189–203.PubMed
141.
go back to reference Rello-Varona S, Vitale I, Kepp O, Senovilla L, Jemaa M, Metivier D, et al. Preferential killing of tetraploid tumor cells by targeting the mitotic kinesin eg5. Cell Cycle. 2009;8:1030–5.PubMed Rello-Varona S, Vitale I, Kepp O, Senovilla L, Jemaa M, Metivier D, et al. Preferential killing of tetraploid tumor cells by targeting the mitotic kinesin eg5. Cell Cycle. 2009;8:1030–5.PubMed
Metadata
Title
Radiation-induced cell death mechanisms
Authors
David Eriksson
Torgny Stigbrand
Publication date
01-08-2010
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 4/2010
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-010-0042-8

Other articles of this Issue 4/2010

Tumor Biology 4/2010 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine