Skip to main content
Erschienen in: Journal of Cancer Research and Clinical Oncology 11/2017

07.08.2017 | Review – Cancer Research

Role of autophagy in regulating the radiosensitivity of tumor cells

verfasst von: Yong Xin, Fan Jiang, Chunsheng Yang, Qiuyue Yan, Wenwen Guo, Qian Huang, Longzhen Zhang, Guan Jiang

Erschienen in: Journal of Cancer Research and Clinical Oncology | Ausgabe 11/2017

Einloggen, um Zugang zu erhalten

Abstract

Background

Autophagy is a metabolic response of cells to chemical and physical factors, such as nutrition or growth factor deprivation, proinflammatory state, hypoxia, accumulation of reactive oxygen species, presence of infectious agents, and DNA damage. Autophagy maintains the homeostasis of intracellular metabolism mainly by degrading cellular organelles and critical proteins. In a sense, autophagy protects cells from death. Radiotherapy is a powerful tool used to control tumor growth, and it can induce autophagy. The relationship between radiotherapy and autophagy is worthy of further investigation.

Methods

We searched various electronic databases including PubMed for peer-reviewed English-language articles and selected articles on the mechanism of autophagy, its role in cancer development and cancer treatment, and the relationship between the effect of radiation therapy and autophagy intensity.

Results

This review has recently shown that the sensitivity of tumor cells to radiation therapy can be increased by regulating autophagy.

Conclusion

The effects of autophagy vary, and autophagy provides various ways of enhancing radiosensitivity, including inhibition of autophagy, increase in autophagy, and altering the outcome of autophagy.
Literatur
Zurück zum Zitat Albert JM, Cao C, Kim KW et al (2007) Inhibition of poly (ADP-ribose) polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models. Clin Cancer Res 13(10):3033–3042PubMedCrossRef Albert JM, Cao C, Kim KW et al (2007) Inhibition of poly (ADP-ribose) polymerase enhances cell death and improves tumor growth delay in irradiated lung cancer models. Clin Cancer Res 13(10):3033–3042PubMedCrossRef
Zurück zum Zitat Alotaibi M, Sharma K, Saleh T et al (2016) Radiosensitization by PARP inhibition in DNA repair proficient and Deficient tumor cells: proliferative recovery in senescent cells. Radiat Res 185(3):229–245PubMedPubMedCentralCrossRef Alotaibi M, Sharma K, Saleh T et al (2016) Radiosensitization by PARP inhibition in DNA repair proficient and Deficient tumor cells: proliferative recovery in senescent cells. Radiat Res 185(3):229–245PubMedPubMedCentralCrossRef
Zurück zum Zitat Altmeyer A, Josset E, Denis JM et al (2012) The mTOR inhibitor RAD001 augments radiation-induced growth inhibition in a hepatocellular carcinoma cell line by increasing autophagy. Int J Oncol 41(4):1381–1386PubMedCrossRef Altmeyer A, Josset E, Denis JM et al (2012) The mTOR inhibitor RAD001 augments radiation-induced growth inhibition in a hepatocellular carcinoma cell line by increasing autophagy. Int J Oncol 41(4):1381–1386PubMedCrossRef
Zurück zum Zitat Anbalagan S, Pires IM, Blick C et al (2012) Radiosensitization of renal cell carcinoma in vitro through the induction of autophagy. Radiother Oncol 103(3):388–393PubMedCrossRef Anbalagan S, Pires IM, Blick C et al (2012) Radiosensitization of renal cell carcinoma in vitro through the induction of autophagy. Radiother Oncol 103(3):388–393PubMedCrossRef
Zurück zum Zitat Apel A, Herr I, Schwarz H et al (2008) Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Can Res 68(5):1485–1494CrossRef Apel A, Herr I, Schwarz H et al (2008) Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Can Res 68(5):1485–1494CrossRef
Zurück zum Zitat Benbrook DM, Long A (2012) Integration of autophagy, proteasomal degradation, unfolded protein response and apoptosis. Exp Oncol 34(3):286–297PubMed Benbrook DM, Long A (2012) Integration of autophagy, proteasomal degradation, unfolded protein response and apoptosis. Exp Oncol 34(3):286–297PubMed
Zurück zum Zitat Bristol ML, Emery SM, Maycotte P et al (2013) Autophagy inhibition for chemosensitization and radiosensitization in cancer: do the preclinical data support this therapeutic strategy? J Pharmacol Exp Ther 344(3):544–552PubMedPubMedCentralCrossRef Bristol ML, Emery SM, Maycotte P et al (2013) Autophagy inhibition for chemosensitization and radiosensitization in cancer: do the preclinical data support this therapeutic strategy? J Pharmacol Exp Ther 344(3):544–552PubMedPubMedCentralCrossRef
Zurück zum Zitat Cao C, Subhawong T, Albert JM et al (2006) Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Can Res 66(20):10040–10047CrossRef Cao C, Subhawong T, Albert JM et al (2006) Inhibition of mammalian target of rapamycin or apoptotic pathway induces autophagy and radiosensitizes PTEN null prostate cancer cells. Can Res 66(20):10040–10047CrossRef
Zurück zum Zitat Chaachouay H, Ohneseit P, Toulany M et al (2011) Autophagy contributes to resistance of tumor cells to ionizing radiation. Radiother Oncol 99(3):287–292PubMedCrossRef Chaachouay H, Ohneseit P, Toulany M et al (2011) Autophagy contributes to resistance of tumor cells to ionizing radiation. Radiother Oncol 99(3):287–292PubMedCrossRef
Zurück zum Zitat Chang L, Graham PH, Hao J et al (2014) PI3K/Akt/mTOR pathway inhibitors enhance radiosensitivity in radioresistant prostate cancer cells through inducing apoptosis, reducing autophagy, suppressing NHEJ and HR repair pathways. Cell Death Dis 5(10):e1437PubMedPubMedCentralCrossRef Chang L, Graham PH, Hao J et al (2014) PI3K/Akt/mTOR pathway inhibitors enhance radiosensitivity in radioresistant prostate cancer cells through inducing apoptosis, reducing autophagy, suppressing NHEJ and HR repair pathways. Cell Death Dis 5(10):e1437PubMedPubMedCentralCrossRef
Zurück zum Zitat Chen N, Karantza-Wadsworth V (2009) Role and regulation of autophagy in cancer. Biochimica et Biophysica Acta (BBA)-Mol Cell Res 1793(9):1516–1523CrossRef Chen N, Karantza-Wadsworth V (2009) Role and regulation of autophagy in cancer. Biochimica et Biophysica Acta (BBA)-Mol Cell Res 1793(9):1516–1523CrossRef
Zurück zum Zitat Chen S, Rehman SK, Zhang W et al (2010) Autophagy is a therapeutic target in anticancer drug resistance. Biochimica et Biophysica Acta (BBA)-Rev Cancer 1806(2):220–229CrossRef Chen S, Rehman SK, Zhang W et al (2010) Autophagy is a therapeutic target in anticancer drug resistance. Biochimica et Biophysica Acta (BBA)-Rev Cancer 1806(2):220–229CrossRef
Zurück zum Zitat Chen YS, Song HX, Lu Y et al (2011) Autophagy inhibition contributes to radiation sensitization of esophageal squamous carcinoma cells. Dis Esophagus 24(6):437–443PubMedCrossRef Chen YS, Song HX, Lu Y et al (2011) Autophagy inhibition contributes to radiation sensitization of esophageal squamous carcinoma cells. Dis Esophagus 24(6):437–443PubMedCrossRef
Zurück zum Zitat Chen Y, Fu LL, Wen X et al (2014a) Oncogenic and tumor suppressive roles of microRNAs in apoptosis and autophagy. Apoptosis 19(8):1177–1189PubMedCrossRef Chen Y, Fu LL, Wen X et al (2014a) Oncogenic and tumor suppressive roles of microRNAs in apoptosis and autophagy. Apoptosis 19(8):1177–1189PubMedCrossRef
Zurück zum Zitat Chen N, Wu L, Yuan H et al (2014b) ROS/Autophagy/Nrf2 pathway mediated low-dose radiation induced radio-resistance in human lung adenocarcinoma A549 cell. Int J Biol Sci 11(7):833–844CrossRef Chen N, Wu L, Yuan H et al (2014b) ROS/Autophagy/Nrf2 pathway mediated low-dose radiation induced radio-resistance in human lung adenocarcinoma A549 cell. Int J Biol Sci 11(7):833–844CrossRef
Zurück zum Zitat Chen Y, Li X, Guo L et al (2015) Combining radiation with autophagy inhibition enhances suppression of tumor growth and angiogenesis in esophageal cancer. Mol Med Rep 12(2):1645–1652PubMedPubMedCentralCrossRef Chen Y, Li X, Guo L et al (2015) Combining radiation with autophagy inhibition enhances suppression of tumor growth and angiogenesis in esophageal cancer. Mol Med Rep 12(2):1645–1652PubMedPubMedCentralCrossRef
Zurück zum Zitat Cheng H, Li J, Liu C et al (2013) Profilin1 sensitizes pancreatic cancer cells to irradiation by inducing apoptosis and reducing autophagy. Curr Mol Med 13(8):1368–1375PubMedCrossRef Cheng H, Li J, Liu C et al (2013) Profilin1 sensitizes pancreatic cancer cells to irradiation by inducing apoptosis and reducing autophagy. Curr Mol Med 13(8):1368–1375PubMedCrossRef
Zurück zum Zitat Chiang PC, Lin SC, Pan SL et al (2010) Antroquinonol displays anticancer potential against human hepatocellular carcinoma cells: a crucial role of AMPK and mTOR pathways. Biochem Pharmacol 79(2):162–171PubMedCrossRef Chiang PC, Lin SC, Pan SL et al (2010) Antroquinonol displays anticancer potential against human hepatocellular carcinoma cells: a crucial role of AMPK and mTOR pathways. Biochem Pharmacol 79(2):162–171PubMedCrossRef
Zurück zum Zitat Chiu HW, Ho SY, Guo HR et al (2009) Combination treatment with arsenic trioxide and irradiation enhances autophagic effects in U118-MG cells through increased mitotic arrest and regulation of PI3K/Akt and ERK1/2 signaling pathways. Autophagy 5(4):472–483PubMedCrossRef Chiu HW, Ho SY, Guo HR et al (2009) Combination treatment with arsenic trioxide and irradiation enhances autophagic effects in U118-MG cells through increased mitotic arrest and regulation of PI3K/Akt and ERK1/2 signaling pathways. Autophagy 5(4):472–483PubMedCrossRef
Zurück zum Zitat Chiu HW, Lin JH, Chen YA et al (2010) Combination treatment with arsenic trioxide and irradiation enhances cell-killing effects in human fibrosarcoma cells in vitro and in vivo through induction of both autophagy and apoptosis. Autophagy 6(3):353–365PubMedCrossRef Chiu HW, Lin JH, Chen YA et al (2010) Combination treatment with arsenic trioxide and irradiation enhances cell-killing effects in human fibrosarcoma cells in vitro and in vivo through induction of both autophagy and apoptosis. Autophagy 6(3):353–365PubMedCrossRef
Zurück zum Zitat Chiu HW, Chen YA, Ho SY et al (2012a) Arsenic trioxide enhances the radiation sensitivity of androgen-dependent and-independent human prostate cancer cells. PLoS One 7(2):e31579PubMedPubMedCentralCrossRef Chiu HW, Chen YA, Ho SY et al (2012a) Arsenic trioxide enhances the radiation sensitivity of androgen-dependent and-independent human prostate cancer cells. PLoS One 7(2):e31579PubMedPubMedCentralCrossRef
Zurück zum Zitat Chiu HW, Fang WH, Chen YL et al (2012b) Monascuspiloin enhances the radiation sensitivity of human prostate cancer cells by stimulating endoplasmic reticulum stress and inducing autophagy. PLoS One 7(7):e40462PubMedPubMedCentralCrossRef Chiu HW, Fang WH, Chen YL et al (2012b) Monascuspiloin enhances the radiation sensitivity of human prostate cancer cells by stimulating endoplasmic reticulum stress and inducing autophagy. PLoS One 7(7):e40462PubMedPubMedCentralCrossRef
Zurück zum Zitat Chiu HW, Yeh YL, Wang YC et al (2013) Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, enhances radiosensitivity and suppresses lung metastasis in breast cancer in vitro and in vivo. PLoS One 8(10):e76340PubMedPubMedCentralCrossRef Chiu HW, Yeh YL, Wang YC et al (2013) Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, enhances radiosensitivity and suppresses lung metastasis in breast cancer in vitro and in vivo. PLoS One 8(10):e76340PubMedPubMedCentralCrossRef
Zurück zum Zitat Chiu HW, Lin SW, Lin LC et al (2015) Synergistic antitumor effects of radiation and proteasome inhibitor treatment in pancreatic cancer through the induction of autophagy and the downregulation of TRAF6. Cancer Lett 365(2):229–239PubMedCrossRef Chiu HW, Lin SW, Lin LC et al (2015) Synergistic antitumor effects of radiation and proteasome inhibitor treatment in pancreatic cancer through the induction of autophagy and the downregulation of TRAF6. Cancer Lett 365(2):229–239PubMedCrossRef
Zurück zum Zitat Choi EJ, Ryu YK, Kim SY et al (2010) Targeting epidermal growth factor receptor–associated signaling pathways in non–small cell lung cancer cells: implication in radiation response. Mol Cancer Res 8(7):1027–1036PubMedCrossRef Choi EJ, Ryu YK, Kim SY et al (2010) Targeting epidermal growth factor receptor–associated signaling pathways in non–small cell lung cancer cells: implication in radiation response. Mol Cancer Res 8(7):1027–1036PubMedCrossRef
Zurück zum Zitat Daido S, Yamamoto A, Fujiwara K et al (2005) Inhibition of the DNA-dependent protein kinase catalytic subunit radiosensitizes malignant glioma cells by inducing autophagy. Can Res 65(10):4368–4375CrossRef Daido S, Yamamoto A, Fujiwara K et al (2005) Inhibition of the DNA-dependent protein kinase catalytic subunit radiosensitizes malignant glioma cells by inducing autophagy. Can Res 65(10):4368–4375CrossRef
Zurück zum Zitat Dumont FJ, Bischoff P (2012) Disrupting the mTOR signaling network as a potential strategy for the enhancement of cancer radiotherapy. Curr Cancer Drug Targets 12(8):899–924PubMedCrossRef Dumont FJ, Bischoff P (2012) Disrupting the mTOR signaling network as a potential strategy for the enhancement of cancer radiotherapy. Curr Cancer Drug Targets 12(8):899–924PubMedCrossRef
Zurück zum Zitat Emery SM, Alotaibi MR, Tao Q et al (2014) Combined antiproliferative effects of the aminoalkylindole WIN55, 212-2 and radiation in breast cancer cells. J Pharmacol Exp Ther 348(2):293–302PubMedPubMedCentralCrossRef Emery SM, Alotaibi MR, Tao Q et al (2014) Combined antiproliferative effects of the aminoalkylindole WIN55, 212-2 and radiation in breast cancer cells. J Pharmacol Exp Ther 348(2):293–302PubMedPubMedCentralCrossRef
Zurück zum Zitat Frankel LB, Lund AH (2012) MicroRNA regulation of autophagy. Carcinogenesis 33(11):2018–2025PubMedCrossRef Frankel LB, Lund AH (2012) MicroRNA regulation of autophagy. Carcinogenesis 33(11):2018–2025PubMedCrossRef
Zurück zum Zitat Gewirtz DA (2007) Autophagy as a mechanism of radiation sensitization in breast tumor cells. Autophagy 3(3):249–250PubMedCrossRef Gewirtz DA (2007) Autophagy as a mechanism of radiation sensitization in breast tumor cells. Autophagy 3(3):249–250PubMedCrossRef
Zurück zum Zitat Gewirtz DA, Hilliker ML, Wilson EN (2009) Promotion of autophagy as a mechanism for radiation sensitization of breast tumor cells. Radiother Oncol 92(3):323–328PubMedCrossRef Gewirtz DA, Hilliker ML, Wilson EN (2009) Promotion of autophagy as a mechanism for radiation sensitization of breast tumor cells. Radiother Oncol 92(3):323–328PubMedCrossRef
Zurück zum Zitat Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23(16):2891–2906PubMedCrossRef Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23(16):2891–2906PubMedCrossRef
Zurück zum Zitat Gravina GL, Marampon F, Sherris D et al (2014) Torc1/Torc2 inhibitor, Palomid 529, enhances radiation response modulating CRM1-mediated survivin function and delaying DNA repair in prostate cancer models. Prostate 74(8):852–868PubMedCrossRef Gravina GL, Marampon F, Sherris D et al (2014) Torc1/Torc2 inhibitor, Palomid 529, enhances radiation response modulating CRM1-mediated survivin function and delaying DNA repair in prostate cancer models. Prostate 74(8):852–868PubMedCrossRef
Zurück zum Zitat Han MW, Lee JC, Choi JY et al (2014) Autophagy inhibition can overcome radioresistance in breast cancer cells through suppression of TAK1 activation. Anticancer Res 34(3):1449–1455PubMed Han MW, Lee JC, Choi JY et al (2014) Autophagy inhibition can overcome radioresistance in breast cancer cells through suppression of TAK1 activation. Anticancer Res 34(3):1449–1455PubMed
Zurück zum Zitat He JH, Liao XL, Wang W et al (2014a) Apogossypolone, a small-molecule inhibitor of Bcl-2, induces radiosensitization of nasopharyngeal carcinoma cells by stimulating autophagy. Int J Oncol 45(3):1099–1108PubMedCrossRef He JH, Liao XL, Wang W et al (2014a) Apogossypolone, a small-molecule inhibitor of Bcl-2, induces radiosensitization of nasopharyngeal carcinoma cells by stimulating autophagy. Int J Oncol 45(3):1099–1108PubMedCrossRef
Zurück zum Zitat He G, Wang Y, Pang X et al (2014b) Inhibition of autophagy induced by TSA sensitizes colon cancer cell to radiation. Tumor Biology 35(2):1003–1011PubMedCrossRef He G, Wang Y, Pang X et al (2014b) Inhibition of autophagy induced by TSA sensitizes colon cancer cell to radiation. Tumor Biology 35(2):1003–1011PubMedCrossRef
Zurück zum Zitat Hideakiito SD, Kanzawa T, Kondo S et al (2005) Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol 26:1401–1410 Hideakiito SD, Kanzawa T, Kondo S et al (2005) Radiation-induced autophagy is associated with LC3 and its inhibition sensitizes malignant glioma cells. Int J Oncol 26:1401–1410
Zurück zum Zitat Høyer-Hansen M, Jäättelä M (2008) Autophagy: an emerging target for cancer therapy. Autophagy 4(5):574–580PubMedCrossRef Høyer-Hansen M, Jäättelä M (2008) Autophagy: an emerging target for cancer therapy. Autophagy 4(5):574–580PubMedCrossRef
Zurück zum Zitat Jin X, Li F, Zheng X et al (2015) Carbon ions induce autophagy effectively through stimulating the unfolded protein response and subsequent inhibiting Akt phosphorylation in tumor cells. Sci Rep 5:13815. doi:10.1038/srep13815 Jin X, Li F, Zheng X et al (2015) Carbon ions induce autophagy effectively through stimulating the unfolded protein response and subsequent inhibiting Akt phosphorylation in tumor cells. Sci Rep 5:13815. doi:10.​1038/​srep13815
Zurück zum Zitat Karagounis IV, Kalamida D, Mitrakas A et al (2016) Repression of the autophagic response sensitises lung cancer cells to radiation and chemotherapy. Br J Cancer 115(3):312–321PubMedPubMedCentralCrossRef Karagounis IV, Kalamida D, Mitrakas A et al (2016) Repression of the autophagic response sensitises lung cancer cells to radiation and chemotherapy. Br J Cancer 115(3):312–321PubMedPubMedCentralCrossRef
Zurück zum Zitat Kim KW, Hwang M, Moretti L et al (2008a) Autophagy upregulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse model of lung cancer. Autophagy 4(5):659–668PubMedPubMedCentralCrossRef Kim KW, Hwang M, Moretti L et al (2008a) Autophagy upregulation by inhibitors of caspase-3 and mTOR enhances radiotherapy in a mouse model of lung cancer. Autophagy 4(5):659–668PubMedPubMedCentralCrossRef
Zurück zum Zitat Kim KW, Moretti L, Lu B (2008b) M867, a novel selective inhibitor of caspase-3 enhances cell death and extends tumor growth delay in irradiated lung cancer models. PLoS One 3(5):e2275PubMedPubMedCentralCrossRef Kim KW, Moretti L, Lu B (2008b) M867, a novel selective inhibitor of caspase-3 enhances cell death and extends tumor growth delay in irradiated lung cancer models. PLoS One 3(5):e2275PubMedPubMedCentralCrossRef
Zurück zum Zitat Kim EJ, Jeong JH, Bae S et al (2013) mTOR inhibitors radiosensitize PTEN-deficient non-small-cell lung cancer cells harboring an EGFR activating mutation by inducing autophagy. J Cell Biochem 114(6):1248–1256PubMedCrossRef Kim EJ, Jeong JH, Bae S et al (2013) mTOR inhibitors radiosensitize PTEN-deficient non-small-cell lung cancer cells harboring an EGFR activating mutation by inducing autophagy. J Cell Biochem 114(6):1248–1256PubMedCrossRef
Zurück zum Zitat Ko A, Kanehisa A, Martins I et al (2014) Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ 21(1):92–99PubMedCrossRef Ko A, Kanehisa A, Martins I et al (2014) Autophagy inhibition radiosensitizes in vitro, yet reduces radioresponses in vivo due to deficient immunogenic signalling. Cell Death Differ 21(1):92–99PubMedCrossRef
Zurück zum Zitat Koukourakis MI, Kalamida D, Mitrakas A et al (2015) Intensified autophagy compromises the efficacy of radiotherapy against prostate cancer. Biochem Biophys Res Commun 461(2):268–274PubMedCrossRef Koukourakis MI, Kalamida D, Mitrakas A et al (2015) Intensified autophagy compromises the efficacy of radiotherapy against prostate cancer. Biochem Biophys Res Commun 461(2):268–274PubMedCrossRef
Zurück zum Zitat Law BYK, Wang M, Ma DL et al (2010) Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis. Mol Cancer Ther 9(3):718–730PubMedCrossRef Law BYK, Wang M, Ma DL et al (2010) Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis. Mol Cancer Ther 9(3):718–730PubMedCrossRef
Zurück zum Zitat Li X, Cen Y, Cai Y et al (2016) TLR9-ERK-mTOR signaling is critical for autophagic cell death induced by CpG oligodeoxynucleotide 107 combined with irradiation in glioma cells. Sci Rep 6:27104PubMedPubMedCentralCrossRef Li X, Cen Y, Cai Y et al (2016) TLR9-ERK-mTOR signaling is critical for autophagic cell death induced by CpG oligodeoxynucleotide 107 combined with irradiation in glioma cells. Sci Rep 6:27104PubMedPubMedCentralCrossRef
Zurück zum Zitat Liang B, Kong D, Liu Y et al (2012) Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells. Radiat Oncol 7(1):213PubMedPubMedCentralCrossRef Liang B, Kong D, Liu Y et al (2012) Autophagy inhibition plays the synergetic killing roles with radiation in the multi-drug resistant SKVCR ovarian cancer cells. Radiat Oncol 7(1):213PubMedPubMedCentralCrossRef
Zurück zum Zitat Liu M, Ma S, Liu M et al (2014) Synergistic killing of lung cancer cells by cisplatin and radiation via autophagy and apoptosis. Oncol Lett 7(6):1903–1910PubMedPubMedCentral Liu M, Ma S, Liu M et al (2014) Synergistic killing of lung cancer cells by cisplatin and radiation via autophagy and apoptosis. Oncol Lett 7(6):1903–1910PubMedPubMedCentral
Zurück zum Zitat Liu C, He W, Jin M et al (2015) Blockage of autophagy in C6 glioma cells enhanced radiosensitivity possibly by attenuating DNA-PK-dependent DSB due to limited ku nuclear translocation and DNA binding. Curr Mol Med 15(7):663–673PubMedCrossRef Liu C, He W, Jin M et al (2015) Blockage of autophagy in C6 glioma cells enhanced radiosensitivity possibly by attenuating DNA-PK-dependent DSB due to limited ku nuclear translocation and DNA binding. Curr Mol Med 15(7):663–673PubMedCrossRef
Zurück zum Zitat Liu S, Wang X, Lu J et al (2016) Ubenimex enhances the radiosensitivity of renal cell carcinoma cells by inducing autophagic cell death. Oncol Lett 12(5):3403–3410PubMedPubMedCentral Liu S, Wang X, Lu J et al (2016) Ubenimex enhances the radiosensitivity of renal cell carcinoma cells by inducing autophagic cell death. Oncol Lett 12(5):3403–3410PubMedPubMedCentral
Zurück zum Zitat Ma L, Chen Z, Erdjument-Bromage H et al (2005) Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosis and cancer pathogenesis. Cell 121(2):179–193PubMedCrossRef Ma L, Chen Z, Erdjument-Bromage H et al (2005) Phosphorylation and functional inactivation of TSC2 by Erk: implications for tuberous sclerosis and cancer pathogenesis. Cell 121(2):179–193PubMedCrossRef
Zurück zum Zitat Meng MB, Wang HH, Guo WH et al (2015) Targeting pyruvate kinase M2 contributes to radiosensitivity of non-small cell lung cancer cells in vitro and in vivo. Cancer Lett 356(2):985–993PubMedCrossRef Meng MB, Wang HH, Guo WH et al (2015) Targeting pyruvate kinase M2 contributes to radiosensitivity of non-small cell lung cancer cells in vitro and in vivo. Cancer Lett 356(2):985–993PubMedCrossRef
Zurück zum Zitat Mo N, Lu YK, Xie WM et al (2014) Inhibition of autophagy enhances the radiosensitivity of nasopharyngeal carcinoma by reducing Rad51 expression. Oncol Rep 32(5):1905–1912PubMedCrossRef Mo N, Lu YK, Xie WM et al (2014) Inhibition of autophagy enhances the radiosensitivity of nasopharyngeal carcinoma by reducing Rad51 expression. Oncol Rep 32(5):1905–1912PubMedCrossRef
Zurück zum Zitat Moretti L, Yang ES, Kim KW et al (2007) Autophagy signaling in cancer and its potential as novel target to improve anticancer therapy. Drug Resist Updates 10(4):135–143CrossRef Moretti L, Yang ES, Kim KW et al (2007) Autophagy signaling in cancer and its potential as novel target to improve anticancer therapy. Drug Resist Updates 10(4):135–143CrossRef
Zurück zum Zitat Mukubou H, Tsujimura T, Sasaki R et al (2010) The role of autophagy in the treatment of pancreatic cancer with gemcitabine and ionizing radiation. Int J Oncol 37(4):821PubMed Mukubou H, Tsujimura T, Sasaki R et al (2010) The role of autophagy in the treatment of pancreatic cancer with gemcitabine and ionizing radiation. Int J Oncol 37(4):821PubMed
Zurück zum Zitat No M, Choi EJ, Kim IA (2009) Targeting HER2 signaling pathway for radiosensitization: alternative strategy for therapeutic resistance. Cancer Biol Ther 8(24):2351–2361PubMedCrossRef No M, Choi EJ, Kim IA (2009) Targeting HER2 signaling pathway for radiosensitization: alternative strategy for therapeutic resistance. Cancer Biol Ther 8(24):2351–2361PubMedCrossRef
Zurück zum Zitat Ondrej M, Cechakova L, Durisova K et al (2016) To live or let die: unclear task of autophagy in the radiosensitization battle. Radiother Oncol 119(2):265–275PubMedCrossRef Ondrej M, Cechakova L, Durisova K et al (2016) To live or let die: unclear task of autophagy in the radiosensitization battle. Radiother Oncol 119(2):265–275PubMedCrossRef
Zurück zum Zitat Paglin S, Yahalom J (2006) Pathways that regulate autophagy and their role in mediating tumor response to treatment. Autophagy 2(4):291–293PubMedCrossRef Paglin S, Yahalom J (2006) Pathways that regulate autophagy and their role in mediating tumor response to treatment. Autophagy 2(4):291–293PubMedCrossRef
Zurück zum Zitat Palumbo S, Comincini S (2013) Autophagy and ionizing radiation in tumors: the “survive or not survive” dilemma. J Cell Physiol 228(1):1–8PubMedCrossRef Palumbo S, Comincini S (2013) Autophagy and ionizing radiation in tumors: the “survive or not survive” dilemma. J Cell Physiol 228(1):1–8PubMedCrossRef
Zurück zum Zitat Palumbo S, Pirtoli L, Tini P et al (2012) Different involvement of autophagy in human malignant glioma cell lines undergoing irradiation and temozolomide combined treatments. J Cell Biochem 113(7):2308–2318PubMedCrossRef Palumbo S, Pirtoli L, Tini P et al (2012) Different involvement of autophagy in human malignant glioma cell lines undergoing irradiation and temozolomide combined treatments. J Cell Biochem 113(7):2308–2318PubMedCrossRef
Zurück zum Zitat Schaaf MBE, Jutten B, Keulers TG et al (2015) Canonical autophagy does not contribute to cellular radioresistance. Radiother Oncol 114(3):406–412PubMedCrossRef Schaaf MBE, Jutten B, Keulers TG et al (2015) Canonical autophagy does not contribute to cellular radioresistance. Radiother Oncol 114(3):406–412PubMedCrossRef
Zurück zum Zitat Shao CJ, Wu MW, Chen FR et al (2012) Histone deacetylase inhibitor, 2-propylpentanoic acid, increases the chemosensitivity and radiosensitivity of human glioma cell lines in vitro. Chin Med J 125(24):4338–4343PubMed Shao CJ, Wu MW, Chen FR et al (2012) Histone deacetylase inhibitor, 2-propylpentanoic acid, increases the chemosensitivity and radiosensitivity of human glioma cell lines in vitro. Chin Med J 125(24):4338–4343PubMed
Zurück zum Zitat Sharma K, Goehe RW, Di X et al (2014) A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy 10(12):2346–2361PubMedCrossRef Sharma K, Goehe RW, Di X et al (2014) A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy 10(12):2346–2361PubMedCrossRef
Zurück zum Zitat Shrivastava S, Mansure JJ, Almajed W et al (2016) The role of HMGB1 in radioresistance of bladder cancer. Mol Cancer Ther 15(3):471–479PubMedCrossRef Shrivastava S, Mansure JJ, Almajed W et al (2016) The role of HMGB1 in radioresistance of bladder cancer. Mol Cancer Ther 15(3):471–479PubMedCrossRef
Zurück zum Zitat Song H, Pan B, Yi J et al (2014) Autophagic activation with Nimotuzumab enhanced chemosensitivity and radiosensitivity of esophageal squamous cell carcinoma. Exp Biol Med 239(5):529–541CrossRef Song H, Pan B, Yi J et al (2014) Autophagic activation with Nimotuzumab enhanced chemosensitivity and radiosensitivity of esophageal squamous cell carcinoma. Exp Biol Med 239(5):529–541CrossRef
Zurück zum Zitat Su YC, Yu CC, Hsu FT et al (2014) Everolimus sensitizes Ras-transformed cells to radiation in vitro through the autophagy pathway. Int J Mol Med 34(5):1417–1422PubMedCrossRef Su YC, Yu CC, Hsu FT et al (2014) Everolimus sensitizes Ras-transformed cells to radiation in vitro through the autophagy pathway. Int J Mol Med 34(5):1417–1422PubMedCrossRef
Zurück zum Zitat Sun Q, Liu T, Yuan Y et al (2015a) MiR-200c inhibits autophagy and enhances radiosensitivity in breast cancer cells by targeting UBQLN1. Int J Cancer 136(5):1003–1012PubMedCrossRef Sun Q, Liu T, Yuan Y et al (2015a) MiR-200c inhibits autophagy and enhances radiosensitivity in breast cancer cells by targeting UBQLN1. Int J Cancer 136(5):1003–1012PubMedCrossRef
Zurück zum Zitat Sun Q, Liu T, Zhang T et al (2015b) MiR-101 sensitizes human nasopharyngeal carcinoma cells to radiation by targeting stathmin 1. Mol Med Rep 11(5):3330–3336PubMedPubMedCentralCrossRef Sun Q, Liu T, Zhang T et al (2015b) MiR-101 sensitizes human nasopharyngeal carcinoma cells to radiation by targeting stathmin 1. Mol Med Rep 11(5):3330–3336PubMedPubMedCentralCrossRef
Zurück zum Zitat Suzuki K, Ohsumi Y (2007) Molecular machinery of autophagosome formation in yeast, Saccharomyces cerevisiae. FEBS Lett 581(11):2156–2161PubMedCrossRef Suzuki K, Ohsumi Y (2007) Molecular machinery of autophagosome formation in yeast, Saccharomyces cerevisiae. FEBS Lett 581(11):2156–2161PubMedCrossRef
Zurück zum Zitat Tang ZP, Xu YD, Peng JR et al (2012) Mechanism of radiosensitization of 3-methyladenine in human hypopharynx cancer cells. Zhonghua er bi yan hou tou jing wai ke za zhi = Chin J Otorhinolaryngol Head Neck Surg 47(11):937–941 Tang ZP, Xu YD, Peng JR et al (2012) Mechanism of radiosensitization of 3-methyladenine in human hypopharynx cancer cells. Zhonghua er bi yan hou tou jing wai ke za zhi = Chin J Otorhinolaryngol Head Neck Surg 47(11):937–941
Zurück zum Zitat Wang W, Long L, Yang N et al (2013a) NVP-BEZ235, a novel dual PI3K/mTOR inhibitor, enhances the radiosensitivity of human glioma stem cells in vitro. Acta Pharmacol Sin 34(5):681–690PubMedPubMedCentralCrossRef Wang W, Long L, Yang N et al (2013a) NVP-BEZ235, a novel dual PI3K/mTOR inhibitor, enhances the radiosensitivity of human glioma stem cells in vitro. Acta Pharmacol Sin 34(5):681–690PubMedPubMedCentralCrossRef
Zurück zum Zitat Wang P, Zhang L, Chen Z et al (2013b) MicroRNA targets autophagy in pancreatic cancer cells during cancer therapy. Autophagy 9(12):2171–2172PubMedCrossRef Wang P, Zhang L, Chen Z et al (2013b) MicroRNA targets autophagy in pancreatic cancer cells during cancer therapy. Autophagy 9(12):2171–2172PubMedCrossRef
Zurück zum Zitat Wang Y, Yin W, Zhu X (2014a) Blocked autophagy enhances radiosensitivity of nasopharyngeal carcinoma cell line CNE-2 in vitro. Acta Otolaryngol 134(1):105–110PubMedCrossRef Wang Y, Yin W, Zhu X (2014a) Blocked autophagy enhances radiosensitivity of nasopharyngeal carcinoma cell line CNE-2 in vitro. Acta Otolaryngol 134(1):105–110PubMedCrossRef
Zurück zum Zitat Wang FZ, Fei H, Cui YJ et al (2014b) The checkpoint 1 kinase inhibitor LY2603618 induces cell cycle arrest, DNA damage response and autophagy in cancer cells. Apoptosis 19(9):1389–1398PubMedCrossRef Wang FZ, Fei H, Cui YJ et al (2014b) The checkpoint 1 kinase inhibitor LY2603618 induces cell cycle arrest, DNA damage response and autophagy in cancer cells. Apoptosis 19(9):1389–1398PubMedCrossRef
Zurück zum Zitat Wilson EN, Bristol ML, Di X et al (2011) A switch between cytoprotective and cytotoxic autophagy in the radiosensitization of breast tumor cells by chloroquine and vitamin D. Horm Cancer 2(5):272–285PubMedPubMedCentralCrossRef Wilson EN, Bristol ML, Di X et al (2011) A switch between cytoprotective and cytotoxic autophagy in the radiosensitization of breast tumor cells by chloroquine and vitamin D. Horm Cancer 2(5):272–285PubMedPubMedCentralCrossRef
Zurück zum Zitat Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22(2):124–131PubMedCrossRef Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22(2):124–131PubMedCrossRef
Zurück zum Zitat Yang Y, Yang Y, Yang X et al (2015) Autophagy and its function in radiosensitivity. Tumor Biol 36(6):4079–4087CrossRef Yang Y, Yang Y, Yang X et al (2015) Autophagy and its function in radiosensitivity. Tumor Biol 36(6):4079–4087CrossRef
Zurück zum Zitat Yang Y, Sun X, Yang Y et al (2016) Gambogic acid enhances the radiosensitivity of human esophageal cancer cells by inducing reactive oxygen species via targeting Akt/mTOR pathway. Tumor Biol 37(2):1853–1862CrossRef Yang Y, Sun X, Yang Y et al (2016) Gambogic acid enhances the radiosensitivity of human esophageal cancer cells by inducing reactive oxygen species via targeting Akt/mTOR pathway. Tumor Biol 37(2):1853–1862CrossRef
Zurück zum Zitat Yao KC, Komata T, Kondo Y et al (2003) Molecular response of human glioblastoma multiforme cells to ionizing radiation: cell cycle arrest, modulation of cyclin-dependent kinase inhibitors, and autophagy. J Neurosurg 98(2):378–384PubMedCrossRef Yao KC, Komata T, Kondo Y et al (2003) Molecular response of human glioblastoma multiforme cells to ionizing radiation: cell cycle arrest, modulation of cyclin-dependent kinase inhibitors, and autophagy. J Neurosurg 98(2):378–384PubMedCrossRef
Zurück zum Zitat Yu L, Tumati V, Tseng SF et al (2012) DAB2IP regulates autophagy in prostate cancer in response to combined treatment of radiation and a DNA-PKcs inhibitor. Neoplasia 14(12):1203–1236PubMedPubMedCentralCrossRef Yu L, Tumati V, Tseng SF et al (2012) DAB2IP regulates autophagy in prostate cancer in response to combined treatment of radiation and a DNA-PKcs inhibitor. Neoplasia 14(12):1203–1236PubMedPubMedCentralCrossRef
Zurück zum Zitat Yu L, Shang ZF, Hsu FM et al (2015) NSCLC cells demonstrate differential mode of cell death in response to the combined treatment of radiation and a DNA-PKcs inhibitor. Oncotarget 6(6):3848–3860PubMedPubMedCentralCrossRef Yu L, Shang ZF, Hsu FM et al (2015) NSCLC cells demonstrate differential mode of cell death in response to the combined treatment of radiation and a DNA-PKcs inhibitor. Oncotarget 6(6):3848–3860PubMedPubMedCentralCrossRef
Zurück zum Zitat Yuan X, Du J, Hua S et al (2015) Suppression of autophagy augments the radiosensitizing effects of STAT3 inhibition on human glioma cells. Exp Cell Res 330(2):267–276PubMedCrossRef Yuan X, Du J, Hua S et al (2015) Suppression of autophagy augments the radiosensitizing effects of STAT3 inhibition on human glioma cells. Exp Cell Res 330(2):267–276PubMedCrossRef
Zurück zum Zitat Zhang C, Qiu X (2015) Andrographolide radiosensitizes human ovarian cancer SKOV3 xenografts due to an enhanced apoptosis and autophagy. Tumor Biol 36(11):8359–8365CrossRef Zhang C, Qiu X (2015) Andrographolide radiosensitizes human ovarian cancer SKOV3 xenografts due to an enhanced apoptosis and autophagy. Tumor Biol 36(11):8359–8365CrossRef
Zurück zum Zitat Zhang X, Shi H, Lin S et al (2015) MicroRNA-216a enhances the radiosensitivity of pancreatic cancer cells by inhibiting beclin-1-mediated autophagy. Oncol Rep 34(3):1557–1564PubMedCrossRef Zhang X, Shi H, Lin S et al (2015) MicroRNA-216a enhances the radiosensitivity of pancreatic cancer cells by inhibiting beclin-1-mediated autophagy. Oncol Rep 34(3):1557–1564PubMedCrossRef
Zurück zum Zitat Zhuang W, Qin Z, Liang Z (2009) The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim Biophys Sin 41(5):341–351PubMedCrossRef Zhuang W, Qin Z, Liang Z (2009) The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim Biophys Sin 41(5):341–351PubMedCrossRef
Zurück zum Zitat Zhuang W, Li B, Long L et al (2011) Knockdown of the DNA-dependent protein kinase catalytic subunit radiosensitizes glioma-initiating cells by inducing autophagy. Brain Res 1371:7–15PubMedCrossRef Zhuang W, Li B, Long L et al (2011) Knockdown of the DNA-dependent protein kinase catalytic subunit radiosensitizes glioma-initiating cells by inducing autophagy. Brain Res 1371:7–15PubMedCrossRef
Metadaten
Titel
Role of autophagy in regulating the radiosensitivity of tumor cells
verfasst von
Yong Xin
Fan Jiang
Chunsheng Yang
Qiuyue Yan
Wenwen Guo
Qian Huang
Longzhen Zhang
Guan Jiang
Publikationsdatum
07.08.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Cancer Research and Clinical Oncology / Ausgabe 11/2017
Print ISSN: 0171-5216
Elektronische ISSN: 1432-1335
DOI
https://doi.org/10.1007/s00432-017-2487-2

Weitere Artikel der Ausgabe 11/2017

Journal of Cancer Research and Clinical Oncology 11/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.