DNA Damage and Repair in Glioblastoma: Emerging Therapeutic Perspectives
https://doi.org/10.24060/2076-3093-2025-15-2-28-42
Abstract
Aggressive and therapy-resistant glioblastoma is among the most lethal malignant tumors in humans. Complete surgical resection is often unachievable; therefore, combination chemoradiotherapy is used to target tumor cells residual beyond the resection margin. This approach induces DNA damage in tumor cells and activates the apoptosis pathway. Unfortunately, recurrence remains a major clinical challenge, frequently manifesting as more aggressive and treatmentresistant glioblastoma phenotypes. The DNA repair and damage response (DDR) pathways are critical for maintaining genome stability. While defects in these mechanisms contribute to oncogenesis, they also make tumor cells vulnerable to DNA-damaging therapy, as the cells become dependent on residual repair capacity. It is of paramount importance to understand the molecular components of these mechanisms and to identify potential therapeutic/pharmacological targets for improving outcomes in glioblastoma patients. A subpopulation of stem-like cells, designated as glioblastoma cancer stem cells (CSCs), has been identified as a critical factor in the initiation, maintenance, and recurrence of tumors. These cells exhibit therapy resistance due to enhanced DNA repair capacity. In addition, emerging evidence suggests a link between carbohydrate metabolism and DNA repair pathways, thereby revealing novel therapeutic vulnerabilities in glioblastoma. This review examines current strategies targeting DNA repair mechanisms in glioblastoma. We present a synopsis of recent advancements in research concerning the mechanisms and factors involved in the elimination of DNA damage induced by ionizing radiation and temozolomide (TMZ). Furthermore, we explore the potential of DNA repair pathway inhibitors under investigation in preclinical and clinical trials.
About the Authors
I. F. GareevRussian Federation
Ilgiz F. Gareev — Cand. Sci. (Med.), Senior Researcher
Ufa; Moscow
O. A. Beylerli
Russian Federation
Ozal A. Beylerli — Cand. Sci. (Med.), Senior Researcher
Moscow
S. A. Roumiantsev
Russian Federation
Sergey A. Roumiantsev — Dr. Sci. (Med.), Prof., Corresponding Member of the Russian Academy of Sciences
Moscow
References
1. Roda D., Veiga P., Melo J.B., Carreira I.M., Ribeiro I.P. Principles in the Management of Glioblastoma. Genes (Basel). 2024;15(4):501. DOI: 10.3390/genes15040501
2. Read R.D., Tapp Z.M., Rajappa P., Hambardzumyan D. Glioblastoma microenvironment-from biology to therapy. Genes Dev. 2024;38(9– 10):360–79. DOI: 10.1101/gad.351427.123
3. Németh E., Szüts D. The mutagenic consequences of defective DNA repair. DNA Repair (Amst). 2024;139:103694. DOI: 10.1016/j.dnarep.2024.103694
4. Hopkins J.L., Lan L., Zou L. DNA repair defects in cancer and therapeutic opportunities. Genes Dev. 2022;36(5–6):278–93. DOI: 10.1101/gad.349431.122
5. Ikliptikawati D.K., Hirai N., Makiyama K., Sabit H., Kinoshita M., Matsumoto K., et al. Nuclear transport surveillance of p53 by nuclear pores in glioblastoma. Cell Rep. 2023;42(8):112882. DOI: 10.1016/j.celrep.2023.112882
6. Le Rhun E., Preusser M., Roth P., Reardon D.A., van den Bent M., Wen P., et al. Molecular targeted therapy of glioblastoma. Cancer Treat Rev. 2019;80:101896. DOI: 10.1016/j.ctrv.2019.101896
7. Butler M., Pongor L., Su Y.T., Xi L., Raffeld M., Quezado M., et al. MGMT Status as a clinical biomarker in glioblastoma. Trends Cancer. 2020;6(5):380–91. DOI: 10.1016/j.trecan.2020.02.010
8. Hashemi M., Etemad S., Rezaei S., Ziaolhagh S., Rajabi R., Rahmanian P., et al. Progress in targeting PTEN/PI3K/Akt axis in glioblastoma therapy: Revisiting molecular interactions. Biomed Pharmacother. 2023;158:114204. DOI: 10.1016/j.biopha.2022.114204
9. Oksenych V., Kainov D.E. DNA Damage Response. Biomolecules. 2021;11(1):123. DOI: 10.3390/biom11010123
10. Chappidi N., Quail T., Doll S., Vogel L.T., Aleksandrov R., Felekyan S., et al. PARP1-DNA co-condensation drives DNA repair site assembly to prevent disjunction of broken DNA ends. Cell. 2024;187(4):945–61. e18. DOI: 10.1016/j.cell.2024.01.015
11. Cheng B., Pan W., Xing Y., Xiao Y., Chen J., Xu Z. Recent advances in DDR (DNA damage response) inhibitors for cancer therapy. Eur J Med Chem. 2022;230:114109. DOI: 10.1016/j.ejmech.2022.114109
12. Chatterjee N., Walker G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ Mol Mutagen. 2017;58(5):235–63. DOI: 10.1002/em.22087
13. Jurkovicova D., Neophytou C.M., Gašparović A.Č., Gonçalves A.C. DNA Damage Response in Cancer Therapy and Resistance: Challenges and Opportunities. Int J Mol Sci. 2022;23(23):14672. DOI: 10.3390/ijms232314672
14. Alghoul E., Basbous J., Constantinou A. Compartmentalization of the DNA damage response: Mechanisms and functions. DNA Repair (Amst). 2023;128:103524. DOI: 10.1016/j.dnarep.2023.103524
15. Vernì F. DNA damage response (DDR) and DNA repair. Int J Mol Sci. 2022;23(13):7204. DOI: 10.3390/ijms23137204
16. Wu L., Sowers J.R., Zhang Y., Ren J. Targeting DNA damage response in cardiovascular diseases: from pathophysiology to therapeutic implications. Cardiovasc Res. 2023;119(3):691–709. DOI: 10.1093/cvr/cvac080
17. Sareen H., Ma Y., Becker T.M., Roberts T.L., de Souza P., Powter B. Molecular Biomarkers in Glioblastoma: A Systematic Review and MetaAnalysis. Int J Mol Sci. 2022;23(16):8835. DOI: 10.3390/ijms23168835
18. Lee Y.C., Lee Y.L., Li C.Y. BRCA Genes and Related Cancers: A MetaAnalysis from Epidemiological Cohort Studies. Medicina (Kaunas). 2021;57(9):905. DOI: 10.3390/medicina57090905
19. Jing X., Yang F., Shao C., Wei K., Xie M., Shen H., et al. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol Cancer. 2019;18(1):157. DOI: 10.1186/s12943-019-1089-9
20. Scanlon S.E., Glazer P.M. Multifaceted control of DNA repair pathways by the hypoxic tumor microenvironment. DNA Repair (Amst). 2015;32:180–9. DOI: 10.1016/j.dnarep.2015.04.030
21. Li M., Thorne R.F., Shi R., Zhang X.D., Li J., Li J., et al. DDIT3 directs a dual mechanism to balance glycolysis and oxidative phosphorylation during glutamine deprivation. Adv Sci (Weinh). 2021;8(11):e2003732. DOI: 10.1002/advs.202003732
22. Tran T.Q., Ishak Gabra M.B., Lowman X.H., Yang Y., Reid M.A., Pan M., et al. Glutamine deficiency induces DNA alkylation damage and sensitizes cancer cells to alkylating agents through inhibition of ALKBH enzymes. PLoS Biol. 2017;15(11):e2002810. DOI: 10.1371/journal.pbio.2002810
23. Goldstein M., Kastan M.B. The DNA damage response: implications for tumor responses to radiation and chemotherapy. Annu Rev Med. 2015;66:129–43. DOI: 10.1146/annurev-med-081313-121208
24. Shaw R., Basu M., Karmakar S., Ghosh M.K. MGMT in TMZ-based glioma therapy: Multifaceted insights and clinical trial perspectives. Biochim Biophys Acta Mol Cell Res. 2024;1871(3):119673. DOI: 10.1016/j.bbamcr.2024.119673
25. Brawanski K.R., Sprung S., Freyschlag C.F., Hoeftberger R., Ströbel T., Haybaeck J., et al. Influence of MMR, MGMT promotor methylation and protein expression on overall and progression-free survival in primary glioblastoma patients treated with temozolomide. Int J Mol Sci. 2023;24(7):6184. DOI: 10.3390/ijms24076184
26. Fahrer J., Christmann M. DNA Alkylation Damage by Nitrosamines and Relevant DNA Repair Pathways. Int J Mol Sci. 2023;24(5):4684. DOI: 10.3390/ijms24054684
27. Tang J.B., Svilar D., Trivedi R.N., Wang X.H., Goellner E.M., Moore B., et al. N-methylpurine DNA glycosylase and DNA polymerase beta modulate BER inhibitor potentiation of glioma cells to temozolomide. Neuro Oncol. 2011;13(5):471–86. DOI: 10.1093/neuonc/nor011
28. Liu J., Bi K., Yang R., Li H., Nikitaki Z., Chang L. Role of DNA damage and repair in radiation cancer therapy: a current update and a look to the future. Int J Radiat Biol. 2020;96(11):1329–38. DOI: 10.1080/09553002.2020.1807641
29. Ghosh S., Ghosh A. Activation of DNA damage response signaling in mammalian cells by ionizing radiation. Free Radic Res. 2021;55(5):581–94. DOI: 10.1080/10715762.2021.1876853
30. Carusillo A., Mussolino C. DNA damage: from threat to treatment. Cells. 2020;9(7):1665. DOI: 10.3390/cells9071665
31. Choi J.E., Chung W.H. Synthetic lethal interaction between oxidative stress response and DNA damage repair in the budding yeast and its application to targeted anticancer therapy. J Microbiol. 2019;57(1):9– 17. DOI: 10.1007/s12275-019-8475-2
32. Vitale I., Kroemer G. Spontaneous DNA damage propels tumorigenicity. Cell Res. 2017;27(6):720–1. DOI: 10.1038/cr.2017.43
33. Graziano S., Gonzalo S. Mechanisms of oncogene-induced genomic instability. Biophys Chem. 2017;225:49–57. DOI: 10.1016/j.bpc.2016.11.008
34. Campisi J. Aging, cellular senescence, and cancer. Annu Rev Physiol. 2013;75:685–705. DOI: 10.1146/annurev-physiol-030212-183653
35. Yabo Y.A., Niclou S.P., Golebiewska A. Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro Oncol. 2022;24(5):669–82. DOI: 10.1093/neuonc/noab269
36. Li C., Qiu S., Liu X., Guo F., Zhai J., Li Z., et al. Extracellular matrixderived mechanical force governs breast cancer cell stemness and quiescence transition through integrin-DDR signaling. Signal Transduct Target Ther. 2023;8(1):247. DOI: 10.1038/s41392-023-01453-0
37. Barzegar Behrooz A., Syahir A., Ahmad S. CD133: beyond a cancer stem cell biomarker. J Drug Target. 2019;27(3):257–69. DOI: 10.1080/1061186X.2018.1479756
38. Min D.W., Kim H.P., Kim J., Wen X., Kim S., Cho Y.W., et al. Phenotype-based single cell sequencing identifies diverse genetic subclones in CD133 positive cancer stem cells. Biochem Biophys Res Commun. 2021;558:209–15. DOI: 10.1016/j.bbrc.2020.09.005
39. Ropolo M., Daga A., Griffero F., Foresta M., Casartelli G., Zunino A., et al. Comparative analysis of DNA repair in stem and nonstem glioma cell cultures. Mol Cancer Res. 2009;7(3):383–92. DOI: 10.1158/1541-7786.MCR-08-0409
40. Bao S., Wu Q., McLendon R.E., Hao Y., Shi Q., Hjelmeland A.B., et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–60. DOI: 10.1038/nature05236
41. Cantidio F.S., Gil G.O.B., Queiroz I.N., Regalin M. Glioblastoma — treatment and obstacles. Rep Pract Oncol Radiother. 2022;27(4):744– 53. DOI: 10.5603/RPOR.a2022.0076
42. Liu G., Yuan X., Zeng Z., Tunici P., Ng H., Abdulkadir I.R., et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 2006;5:67. DOI: 10.1186/1476-4598-5-67
43. Beier D., Schulz J.B., Beier C.P. Chemoresistance of glioblastoma cancer stem cells — much more complex than expected. Mol Cancer. 2011;10:128. DOI: 10.1186/1476-4598-10-128
44. Puigvert J.C., Sanjiv K., Helleday T. Targeting DNA repair, DNA metabolism and replication stress as anti-cancer strategies. FEBS J. 2016;283(2):232–45. DOI: 10.1111/febs.13574
45. Efimova E.V., Takahashi S., Shamsi N.A., Wu D., Labay E., Ulanovskaya O.A., et al. Linking cancer metabolism to DNA repair and accelerated senescence. Mol Cancer Res. 2016;14(2):173–84. DOI: 10.1158/1541-7786.MCR-15-0263
46. Barba I., Carrillo-Bosch L., Seoane J. Targeting the Warburg Effect in Cancer: Where Do We Stand? Int J Mol Sci. 2024;25(6):3142. DOI: 10.3390/ijms25063142
47. Liu X., Li Z., Zhao Q., Zhou X., Wang Y., Zhao G., et al. Capsaicin reverses cisplatin resistance in tongue squamous cell carcinoma by inhibiting the Warburg effect and facilitating mitochondrialdependent apoptosis via the AMPK/AKT/mTOR axis. Cell Biol Int. 2024;48(8):1097–110. DOI: 10.1002/cbin.12169
48. Wen S.S., Wu Y.J., Wang J.Y., Ni Z.X., Dong S., Xie X.J., et al. BRAFV600E/p-ERK/p-DRP1(Ser616) promotes tumor progression and reprogramming of glucose metabolism in papillary thyroid cancer. Thyroid. 2024;34(10):1246–59. DOI: 10.1089/thy.2023.0700
49. Pothuri B., Brodsky A.L., Sparano J.A., Blank S.V., Kim M., Hershman D.L., et al. Phase I and pharmacokinetic study of veliparib, a PARP inhibitor, and pegylated liposomal doxorubicin (PLD) in recurrent gynecologic cancer and triple negative breast cancer with long-term follow-up. Cancer Chemother Pharmacol. 2020;85(4):741–51. DOI: 10.1007/s00280-020-04030-2
50. Dibitetto D., Widmer C.A., Rottenberg S. PARPi, BRCA, and gaps: controversies and future research. Trends Cancer. 2024;10(9):857–69. DOI: 10.1016/j.trecan.2024.06.008
51. Cucchi D., Gibson A., Martin S.A. The emerging relationship between metabolism and DNA repair. Cell Cycle. 2021;20(10):943–59. DOI: 10.1080/15384101.2021.1912889
52. Koo S.Y., Park E.J., Noh H.J., Jo S.M., Ko B.K., Shin H.J., et al. Ubiquitination Links DNA Damage and Repair Signaling to Cancer Metabolism. Int J Mol Sci. 2023;24(9):8441. DOI: 10.3390/ijms24098441
53. Helleday T., Rudd S.G. Targeting the DNA damage response and repair in cancer through nucleotide metabolism. Mol Oncol. 2022;16(21):3792–810. DOI: 10.1002/1878-0261.13227
54. Govoni M., Rossi V., Di Stefano G., Manerba M. Lactate upregulates the expression of DNA repair genes, causing intrinsic resistance of cancer cells to cisplatin. Pathol Oncol Res. 2021;27:1609951. DOI: 10.3389/pore.2021.1609951
55. Fan Z., Ye M., Liu D., Zhou W., Zeng T., He S., et al. Lactate drives the ESM1-SCD1 axis to inhibit the antitumor CD8+ T-cell response by activating the Wnt/β-catenin pathway in ovarian cancer cells and inducing cisplatin resistance. Int Immunopharmacol. 2024;137:112461. DOI: 10.1016/j.intimp.2024.112461
56. Kathagen-Buhmann A., Schulte A., Weller J., Holz M., HeroldMende C., Glass R., et al. Glycolysis and the pentose phosphate pathway are differentially associated with the dichotomous regulation of glioblastoma cell migration versus proliferation. Neuro Oncol. 2016;18(9):1219–29. DOI: 10.1093/neuonc/now024
57. Marin-Valencia I., Cho S.K., Rakheja D., Hatanpaa K.J., Kapur P., Mashimo T., et al. Glucose metabolism via the pentose phosphate pathway, glycolysis and Krebs cycle in an orthotopic mouse model of human brain tumors. NMR Biomed. 2012;25(10):1177–86. DOI: 10.1002/nbm.2787
58. Zhu Z., Kiang K.M., Li N., Liu J., Zhang P., Jin L., et al. Folate enzyme MTHFD2 links one-carbon metabolism to unfolded protein response in glioblastoma. Cancer Lett. 2022;549:215903. DOI: 10.1016/j.canlet.2022.215903
59. Yuen C.A., Asuthkar S., Guda M.R., Tsung A.J., Velpula K.K. Cancer stem cell molecular reprogramming of the Warburg effect in glioblastomas: a new target gleaned from an old concept. CNS Oncol. 2016;5(2):101–8. DOI: 10.2217/cns-2015-0006
60. Mukherjee J., Ohba S., See W.L., Phillips J.J., Molinaro A.M., Pieper R.O. PKM2 uses control of HuR localization to regulate p27 and cell cycle progression in human glioblastoma cells. Int J Cancer. 2016;139(1):99–111. DOI: 10.1002/ijc.30041
61. Goidts V., Bageritz J., Puccio L., Nakata S., Zapatka M., Barbus S., et al. RNAi screening in glioma stem-like cells identifies PFKFB4 as a key molecule important for cancer cell survival. Oncogene. 2012;31(27):3235–43. DOI: 10.1038/onc.2011.490
62. Khan F., Lin Y., Ali H., Pang L., Dunterman M., Hsu W.H., et al. Lactate dehydrogenase A regulates tumor-macrophage symbiosis to promote glioblastoma progression. Nat Commun. 2024;15(1):1987. DOI: 10.1038/s41467-024-46193-z
63. Nguyen T.T.T., Zhang Y., Shang E., Shu C., Torrini C., Zhao J., et al. HDAC inhibitors elicit metabolic reprogramming by targeting superenhancers in glioblastoma models. J Clin Invest. 2020;130(7):3699– 716. DOI: 10.1172/JCI129049
64. Guyon J., Fernandez-Moncada I., Larrieu C.M., Bouchez C.L., Pagano Zottola A.C., Galvis J., et al. Lactate dehydrogenases promote glioblastoma growth and invasion via a metabolic symbiosis. EMBO Mol Med. 2022;14(12):e15343. DOI: 10.15252/emmm.202115343
65. Valvona C.J., Fillmore H.L., Nunn P.B., Pilkington G.J. The Regulation and function of lactate dehydrogenase a: therapeutic potential in brain tumor. Brain Pathol. 2016;26(1):3–17. DOI: 10.1111/bpa.12299
66. Malaquin N., Carrier-Leclerc A., Dessureault M., Rodier F. DDRmediated crosstalk between DNA-damaged cells and their microenvironment. Front Genet. 2015;6:94. DOI: 10.3389/fgene.2015.00094
67. Wang J.Y.J. Cell death response to DNA damage. Yale J Biol Med. 2019;92(4):771–9. PMID: 31866794
68. Bigge J., Koebbe L.L., Giel A.S., Bornholdt D., Buerfent B., Dasmeh P., et al. Expression quantitative trait loci influence DNA damage-induced apoptosis in cancer. BMC Genomics. 2024;25(1):1168. DOI: 10.1186/s12864-024-11068-6
69. Wu S., Li X., Gao F., de Groot J.F., Koul D., Yung W.K.A. PARP-mediated PARylation of MGMT is critical to promote repair of temozolomide-induced O6-methylguanine DNA damage in glioblastoma. Neuro Oncol. 2021;23(6):920–31. DOI: 10.1093/neuonc/noab003
70. Yuan A.L., Meode M., Tan M., Maxwell L., Bering E.A., Pedersen H., et al. PARP inhibition suppresses the emergence of temozolomide resistance in a model system. J Neurooncol. 2020;148(3):463–72. DOI: 10.1007/s11060-020-03561-1
71. Xavier M.A., Rezende F., Titze-de-Almeida R., Cornelissen B. BRCAness as a biomarker of susceptibility to PARP inhibitors in glioblastoma multiforme. Biomolecules. 2021;11(8):1188. DOI: 10.3390/biom11081188
72. Meimand S.E., Pour-Rashidi A., Shahrbabak M.M., Mohammadi E., Meimand F.E., Rezaei N. The prognostication potential of BRCA genes expression in gliomas: a genetic survival analysis study. World Neurosurg. 2022;157:e123–8. DOI: 10.1016/j.wneu.2021.09.107
73. Sun P., Li Y., Chao X., Li J., Luo R., Li M., et al. Clinical characteristics and prognostic implications of BRCA-associated tumors in males: a pan-tumor survey. BMC Cancer. 2020;20(1):994. DOI: 10.1186/s12885-020-07481-1
74. Nile D.L., Rae C., Hyndman I.J., Gaze M.N., Mairs R.J. An evaluation in vitro of PARP-1 inhibitors, rucaparib and olaparib, as radiosensitisers for the treatment of neuroblastoma. BMC Cancer. 2016;16:621. DOI: 10.1186/s12885-016-2656-8
75. Parrish K.E., Cen L., Murray J., Calligaris D., Kizilbash S., Mittapalli R.K., et al. Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther. 2015;14(12):2735–43. DOI: 10.1158/1535-7163.MCT-15-0553
76. van Vuurden D.G., Hulleman E., Meijer O.L., Wedekind L.E., Kool M., Witt H., et al. PARP inhibition sensitizes childhood high grade glioma, medulloblastoma and ependymoma to radiation. Oncotarget. 2011;2(12):984–96. DOI: 10.18632/oncotarget.362
77. Chornenkyy Y., Agnihotri S., Yu M., Buczkowicz P., Rakopoulos P., Golbourn B., et al. Poly-ADP-Ribose polymerase as a therapeutic target in pediatric diffuse intrinsic pontine glioma and pediatric high-grade astrocytoma. Mol Cancer Ther. 2015;14(11):2560–8. DOI: 10.1158/1535-7163.MCT-15-0282
78. Wu S., Gao F., Zheng S., Zhang C., Martinez-Ledesma E., Ezhilarasan R., et al. EGFR amplification induces increased DNA damage response and renders selective sensitivity to talazoparib (PARP Inhibitor) in glioblastoma. Clin Cancer Res. 2020;26(6):1395–407. DOI: 10.1158/1078-0432.CCR-19-2549
79. Sachdev E., Tabatabai R., Roy V., Rimel B.J., Mita M.M. PARP inhibition in cancer: an update on clinical development. Target Oncol. 2019;14(6):657–79. DOI: 10.1007/s11523-019-00680-2
80. Lin F., de Gooijer M.C., Roig E.M., Buil L.C., Christner S.M., Beumer J.H., et al. ABCB1, ABCG2, and PTEN determine the response of glioblastoma to temozolomide and ABT-888 therapy. Clin Cancer Res. 2014;20(10):2703–13. DOI: 10.1158/1078-0432.CCR-14-0084
81. Wagner L.M. Profile of veliparib and its potential in the treatment of solid tumors. Onco Targets Ther. 2015;8:1931–9. DOI: 10.2147/OTT.S69935
82. Barazzuol L., Jena R., Burnet N.G., Meira L.B., Jeynes J.C., Kirkby K.J., et al. Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma. Radiat Oncol. 2013;8:65. DOI: 10.1186/1748-717X-8-65
83. Zhiyu Tang, Bin Jiang, Zhenyan Shi, Wenfeng Gong, Yong Liu, Xing Wang, et al. Abstract 1651: BGB-290, a novel PARP inhibitor with unique brain penetration ability, demonstrated strong synergism with temozolomide in subcutaneous and intracranial xenograft models. Cancer Res. 2015;75 (15_Suppl):1651. DOI:10.1158/1538-7445.AM2015-1651
84. Zimmermann A., Zenke F.T., Chiu L.Y., Dahmen H., Pehl U., Fuchss T., et al. A new class of selective ATM inhibitors as combination partners of DNA double-strand break inducing cancer therapies. Mol Cancer Ther. 2022;21(6):859–70. DOI: 10.1158/1535-7163.MCT-21-0934
85. Priya B., Ravi S., Kirubakaran S. Targeting ATM and ATR for cancer therapeutics: Inhibitors in clinic. Drug Discov Today. 2023;28(8):103662. DOI: 10.1016/j.drudis.2023.103662
86. Manic G., Obrist F., Sistigu A., Vitale I. Trial watch: targeting ATMCHK2 and ATR-CHK1 pathways for anticancer therapy. Mol Cell Oncol. 2015;2(4):e1012976. DOI: 10.1080/23723556.2015.1012976
87. Smith H.L., Southgate H., Tweddle D.A., Curtin N.J. DNA damage checkpoint kinases in cancer. Expert Rev Mol Med. 2020;22:e2. DOI: 10.1017/erm.2020.3
88. Blake S.M., Stricker S.H., Halavach H., Poetsch A.R., Cresswell G., Kelly G., et al. Inactivation of the ATMIN/ATM pathway protects against glioblastoma formation. Elife. 2016;5:e08711. DOI: 10.7554/eLife.08711
89. Vecchio D., Daga A., Carra E., Marubbi D., Raso A., Mascelli S., et al. Pharmacokinetics, pharmacodynamics and efficacy on pediatric tumors of the glioma radiosensitizer KU60019. Int J Cancer. 2015;136(6):1445–57. DOI: 10.1002/ijc.29121
90. Jin M.H., Oh D.Y. ATM in DNA repair in cancer. Pharmacol Ther. 2019;203:107391. DOI: 10.1016/j.pharmthera.2019.07.002
91. Chen J., Laverty D.J., Talele S., Bale A., Carlson B.L., Porath K.A., et al. Aberrant ATM signaling and homology-directed DNA repair as a vulnerability of p53-mutant GBM to AZD1390-mediated radiosensitization. Sci Transl Med. 2024;16(734):eadj5962. DOI: 10.1126/scitranslmed.adj5962
92. Lozinski M., Bowden N.A., Graves M.C., Fay M., Day B.W., Stringer B.W., et al. ATR inhibition using gartisertib enhances cell death and synergises with temozolomide and radiation in patient-derived glioblastoma cell lines. Oncotarget. 2024;15:1–18. DOI: 10.18632/oncotarget.28551
93. Peng C., Chen Z., Wang S., Wang H.W., Qiu W., Zhao L., et al. The error-prone DNA polymerase κ promotes temozolomide resistance in glioblastoma through Rad17-dependent activation of ATR-Chk1 signaling. Cancer Res. 2016;76(8):2340–53. DOI: 10.1158/0008-5472.CAN-15-1884
94. Aasland D., Götzinger L., Hauck L., Berte N., Meyer J., Effenberger M., et al. Temozolomide induces senescence and repression of DNA repair pathways in glioblastoma cells via activation of ATR-CHK1, p21, and NF-κB. Cancer Res. 2019;79(1):99–113. DOI: 10.1158/0008-5472.CAN-18-1733
95. Ganesa S., Sule A., Sundaram R.K., Bindra R.S. Mismatch repair proteins play a role in ATR activation upon temozolomide treatment in MGMT-methylated glioblastoma. Sci Rep. 2022;12(1):5827. DOI: 10.1038/s41598-022-09614-x
96. Chang K.F., Liu C.Y., Huang Y.C., Hsiao C.Y., Tsai N.M. Downregulation of VEGFR2 signaling by cedrol abrogates VEGF-driven angiogenesis and proliferation of glioblastoma cells through AKT/P70S6K and MAPK/ERK1/2 pathways. Oncol Lett. 2023;26(2):342. DOI: 10.3892/ol.2023.13928
97. Gilbert M.R., Dignam J.J., Armstrong T.S., Wefel J.S., Blumenthal D.T., Vogelbaum M.A., et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):699–708. DOI: 10.1056/NEJMoa1308573
98. Carmell N., Rominiyi O., Myers K.N., McGarrity-Cottrell C., Vanderlinden A., Lad N., et al. Identification and validation of ERK5 as a DNA damage modulating drug target in glioblastoma. Cancers (Basel). 2021;13(5):944. DOI: 10.3390/cancers13050944
99. Koncar R.F., Dey B.R., Stanton A.J., Agrawal N., Wassell M.L., McCarl L.H., et al. Identification of novel RAS signaling therapeutic vulnerabilities in diffuse intrinsic pontine gliomas. Cancer Res. 2019;79(16):4026–41. DOI: 10.1158/0008-5472.CAN-18-3521
100. Tripathi S., Najem H., Mahajan A.S., Zhang P., Low J.T., Stegh A.H., et al. cGAS-STING pathway targeted therapies and their applications in the treatment of high-grade glioma. F1000Res. 2022;11:1010. DOI: 10.12688/f1000research.125163.1
101. Low J.T., Brown M.C., Reitman Z.J., Bernstock J.D., Markert J.M., Friedman G.K., et al. Understanding and therapeutically exploiting cGAS/STING signaling in glioblastoma. J Clin Invest. 2024;134(2):e163452. DOI: 10.1172/JCI163452
102. He Y., Yang Y., Huang W., Yang S., Xue X., Zhu K., et al. Manganese facilitated cGAS-STING-IFNI pathway activation induced by ionizing radiation in glioma cells. Int J Radiat Biol. 2023;99(12):1890–907. DOI: 10.1080/09553002.2023.2232011
Review
For citations:
Gareev I.F., Beylerli O.A., Roumiantsev S.A. DNA Damage and Repair in Glioblastoma: Emerging Therapeutic Perspectives. Creative surgery and oncology. 2025;15(2):124-138. (In Russ.) https://doi.org/10.24060/2076-3093-2025-15-2-28-42