Temozolomide, Procarbazine and Nitrosoureas in the Therapy of Malignant Gliomas: Update of Mechanisms, Drug Resistance and Therapeutic Implications
Abstract
:1. Introduction
2. Key Node MGMT
3. Mechanism of Cytotoxicity of O6AA: Upstream Pathways
4. Downstream Events: Signaling Pathways and Apoptosis
5. TMZ-Induced Cellular Senescence
6. Autophagy
7. Are There TMZ Thresholds?
8. Damage Accumulation in MGMT-Lacking Cells
9. How Much O6MeG Is Required for Inducing DSBs, Cell Death, and Senescence?
10. Can TMZ Cause Toxicity in MGMT+ (Promoter-Unmethylated) Cells?
11. TMZ-Mediated Upregulation of MGMT?
12. MGMT Inhibition and Chemoprotection
13. IDH1
14. EGFR Mutation/Amplification
15. Dual Role of p53
16. Synthetic Lethality and Role of PARP
17. Glucocorticoids and TMZ/Procarbazine Therapy
18. Interaction of Radiation with TMZ
19. TMZ and TTF Therapy
20. CUSP9v3 Therapy
21. Immunotherapy (Together with O6AA)
22. CDK Inhibitors
23. Why Are Glioblastomas Refractory to Alkylating Agent-Based Therapy?
24. Can Senescent Cells Become Reactivated to Proliferate?
25. Do We Need Senolytics and Senostatics?
26. Conclusions and Summary
Funding
Acknowledgments
Conflicts of Interest
References
- Tan, A.C.; Ashley, D.M.; Lopez, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of glioblastoma: State of the art and future directions. CA Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef]
- Philips, A.; Henshaw, D.L.; Lamburn, G.; O’Carroll, M.J. Brain Tumours: Rise in Glioblastoma Multiforme Incidence in England 1995–2015 Suggests an Adverse Environmental or Lifestyle Factor. J. Environ. Public Health 2018, 2018, 7910754. [Google Scholar] [CrossRef]
- Low, J.T.; Ostrom, Q.T.; Cioffi, G.; Neff, C.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. Primary brain and other central nervous system tumors in the United States (2014–2018): A summary of the CBTRUS statistical report for clinicians. Neuro-Oncol. Pract. 2022, 9, 165–182. [Google Scholar] [CrossRef] [PubMed]
- Ostrom, Q.T.; Patil, N.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013–2017. Neuro Oncol. 2020, 22, iv1–iv96. [Google Scholar] [CrossRef] [PubMed]
- Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed]
- Wen, P.Y.; Weller, M.; Lee, E.Q.; Alexander, B.M.; Barnholtz-Sloan, J.S.; Barthel, F.P.; Batchelor, T.T.; Bindra, R.S.; Chang, S.M.; Chiocca, E.A.; et al. Glioblastoma in adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020, 22, 1073–1113. [Google Scholar] [CrossRef] [PubMed]
- Margison, G.P.; Kleihues, P. Chemical carcinogenesis in the nervous system. Preferential accumulation of O6-methylguanine in rat brain deoxyribonucleic acid during repetitive administration of N-methyl-N-nitrosourea. Biochem. J. 1975, 148, 521–525. [Google Scholar] [CrossRef] [PubMed]
- Vermel, E.M.; Korman, N.P.; Milonov, B.V.; Evseenko, L.S.; Orlova, R.S. A clinical study of N-nitrosomethylurea. Vopr. Onkol. 1970, 16, 31–37. [Google Scholar] [PubMed]
- Kolaric, K.; Roth, A. Treatment of metastatic brain tumors with the combination of 1-methyl-1-nitrosourea (MNU) and cyclophosphamide. J. Cancer Res. Clin. Oncol. 1980, 97, 193–198. [Google Scholar] [CrossRef]
- Danson, S.J.; Middleton, M.R. Temozolomide: A novel oral alkylating agent. Expert. Rev. Anticancer Ther. 2001, 1, 13–19. [Google Scholar] [CrossRef]
- Weller, M.; van den Bent, M.; Preusser, M.; Le Rhun, E.; Tonn, J.C.; Minniti, G.; Bendszus, M.; Balana, C.; Chinot, O.; Dirven, L.; et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat. Rev. Clin. Oncol. 2021, 18, 170–186. [Google Scholar] [CrossRef]
- Vargas Lopez, A.J. Glioblastoma in adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2021, 23, 502–503. [Google Scholar] [CrossRef]
- Beranek, D.T. Distribution of methyl and ethyl adducts following alkylation with monofunctional alkylating agents. Mutat. Res. 1990, 231, 11–30. [Google Scholar] [CrossRef]
- Stupp, R.; Hegi, M.E.; Gilbert, M.R.; Chakravarti, A. Chemoradiotherapy in malignant glioma: Standard of care and future directions. J. Clin. Oncol. 2007, 25, 4127–4136. [Google Scholar] [CrossRef]
- Strik, H.M.; Marosi, C.; Kaina, B.; Neyns, B. Temozolomide dosing regimens for glioma patients. Curr. Neurol. Neurosci. Rep. 2012, 12, 286–293. [Google Scholar] [CrossRef]
- Herrlinger, U.; Tzaridis, T.; Mack, F.; Steinbach, J.P.; Schlegel, U.; Sabel, M.; Hau, P.; Kortmann, R.D.; Krex, D.; Grauer, O.; et al. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): A randomised, open-label, phase 3 trial. Lancet 2019, 393, 678–688. [Google Scholar] [CrossRef]
- Kaina, B.; Christmann, M.; Naumann, S.; Roos, W.P. MGMT: Key node in the battle against genotoxicity, carcinogenicity and apoptosis induced by alkylating agents. DNA Repair 2007, 6, 1079–1099. [Google Scholar] [CrossRef]
- Christmann, M.; Kaina, B. Epigenetic regulation of DNA repair genes and implications for tumor therapy. Mutat. Res. 2019, 780, 15–28. [Google Scholar] [CrossRef] [PubMed]
- Margison, G.P.; Povey, A.C.; Kaina, B.; Santibanez Koref, M.F. Variability and regulation of O6-alkylguanine-DNA alkyltransferase. Carcinogenesis 2003, 24, 625–635. [Google Scholar] [CrossRef] [PubMed]
- Goth, R.; Rajewsky, M.F. Persistence of O6-ethylguanine in rat-brain DNA: Correlation with nervous system-specific carcinogenesis by ethylnitrosourea. Proc. Natl. Acad. Sci. USA 1974, 71, 639–643. [Google Scholar] [CrossRef] [PubMed]
- Silber, J.R.; Blank, A.; Bobola, M.S.; Mueller, B.A.; Kolstoe, D.D.; Ojemann, G.A.; Berger, M.S. Lack of the DNA repair protein O6-methylguanine-DNA methyltransferase in histologically normal brain adjacent to primary human brain tumors. Proc. Natl. Acad. Sci. USA 1996, 93, 6941–6946. [Google Scholar] [CrossRef]
- Janssen, K.; Eichhorn-Grombacher, U.; Schlink, K.; Nitzsche, S.; Oesch, F.; Kaina, B. Long-time expression of DNA repair enzymes MGMT and APE in human peripheral blood mononuclear cells. Arch. Toxicol. 2001, 75, 306–312. [Google Scholar] [CrossRef]
- Christmann, M.; Kaina, B. Transcriptional regulation of human DNA repair genes following genotoxic stress: Trigger mechanisms, inducible responses and genotoxic adaptation. Nucleic Acids Res. 2013, 41, 8403–8420. [Google Scholar] [CrossRef]
- Wiewrodt, D.; Nagel, G.; Dreimuller, N.; Hundsberger, T.; Perneczky, A.; Kaina, B. MGMT in primary and recurrent human glioblastomas after radiation and chemotherapy and comparison with p53 status and clinical outcome. Int. J. Cancer 2008, 122, 1391–1399. [Google Scholar] [CrossRef]
- Fahrer, J.; Kaina, B. O6-methylguanine-DNA methyltransferase in the defense against N-nitroso compounds and colorectal cancer. Carcinogenesis 2013, 34, 2435–2442. [Google Scholar] [CrossRef]
- Weller, M.; Stupp, R.; Reifenberger, G.; Brandes, A.A.; van den Bent, M.J.; Wick, W.; Hegi, M.E. MGMT promoter methylation in malignant gliomas: Ready for personalized medicine? Nat. Rev. Neurol. 2010, 6, 39–51. [Google Scholar] [CrossRef] [PubMed]
- Mason, M.; Laperriere, N.; Wick, W.; Reardon, D.A.; Malmstrom, A.; Hovey, E.; Weller, M.; Perry, J.R. Glioblastoma in the elderly: Making sense of the evidence. Neuro-Oncol. Pract. 2016, 3, 77–86. [Google Scholar] [CrossRef]
- Ashkan, K.; Baig Mirza, A.; Soumpasis, C.; Syrris, C.; Kalaitzoglou, D.; Sharma, C.; James, Z.J.; Khoja, A.K.; Ahmed, R.; Vastani, A.; et al. MGMT Promoter Methylation: Prognostication beyond Treatment Response. J. Pers. Med. 2023, 13, 999. [Google Scholar] [CrossRef]
- Switzeny, O.J.; Christmann, M.; Renovanz, M.; Giese, A.; Sommer, C.; Kaina, B. MGMT promoter methylation determined by HRM in comparison to MSP and pyrosequencing for predicting high-grade glioma response. Clin. Epigenetics 2016, 8, 49. [Google Scholar] [CrossRef] [PubMed]
- Gerson, S.L. MGMT: Its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer 2004, 4, 296–307. [Google Scholar] [CrossRef] [PubMed]
- Bai, P.; Fan, T.; Sun, G.; Wang, X.; Zhao, L.; Zhong, R. The dual role of DNA repair protein MGMT in cancer prevention and treatment. DNA Repair 2023, 123, 103449. [Google Scholar] [CrossRef] [PubMed]
- Kaina, B.; Fritz, G.; Mitra, S.; Coquerelle, T. Transfection and expression of human O6-methylguanine-DNA methyltransferase (MGMT) cDNA in Chinese hamster cells: The role of MGMT in protection against the genotoxic effects of alkylating agents. Carcinogenesis 1991, 12, 1857–1867. [Google Scholar] [PubMed]
- Yoshioka, K.; Yoshioka, Y.; Hsieh, P. ATR kinase activation mediated by MutSalpha and MutLalpha in response to cytotoxic O6-methylguanine adducts. Mol. Cell 2006, 22, 501–510. [Google Scholar] [CrossRef]
- Fuchs, R.P.; Isogawa, A.; Paulo, J.A.; Onizuka, K.; Takahashi, T.; Amunugama, R.; Duxin, J.P.; Fujii, S. Crosstalk between repair pathways elicits double-strand breaks in alkylated DNA and implications for the action of temozolomide. eLife 2021, 10, e69544. [Google Scholar] [CrossRef]
- Karran, P.; Bignami, M. DNA damage tolerance, mismatch repair and genome instability. Bioassays 1994, 16, 833–839. [Google Scholar] [CrossRef]
- Zhukovskaya, N.; Branch, P.; Aquiliana, G.; Karran, P. DNA replication arrest and tolerance to DNA methylation damage. Carcinogenesis 1994, 15, 2189–2194. [Google Scholar] [CrossRef] [PubMed]
- Quiros, S.; Roos, W.P.; Kaina, B. Processing of O6-methylguanine into DNA double-strand breaks requires two rounds of replication whereas apoptosis is also induced in subsequent cell cycles. Cell Cycle 2010, 9, 168–178. [Google Scholar] [CrossRef]
- Noonan, E.M.; Shah, D.; Yaffe, M.B.; Lauffenburger, D.A.; Samson, L.D. O6-Methylguanine DNA lesions induce an intra-S-phase arrest from which cells exit into apoptosis governed by early and late multi-pathway signaling network activation. Integr. Biol. Quant. Biosci. Nano Macro 2012, 4, 1237–1255. [Google Scholar] [CrossRef]
- Derheimer, F.A.; O’Hagan, H.M.; Krueger, H.M.; Hanasoge, S.; Paulsen, M.T.; Ljungman, M. RPA and ATR link transcriptional stress to p53. Proc. Natl. Acad. Sci. USA 2007, 104, 12778–12783. [Google Scholar] [CrossRef]
- Goldstein, M.; Roos, W.P.; Kaina, B. Apoptotic death induced by the cyclophosphamide analogue mafosfamide in human lymphoblastoid cells: Contribution of DNA replication, transcription inhibition and Chk/p53 signaling. Toxicol. Appl. Pharmacol. 2008, 229, 20–32. [Google Scholar] [CrossRef]
- Roos, W.P.; Kaina, B. DNA damage-induced cell death: From specific DNA lesions to the DNA damage response and apoptosis. Cancer Lett. 2013, 332, 237–248. [Google Scholar] [CrossRef] [PubMed]
- Ochs, K.; Kaina, B. Apoptosis induced by DNA damage O6-methylguanine is Bcl-2 and caspase-9/3 regulated and Fas/caspase-8 independent. Cancer Res. 2000, 60, 5815–5824. [Google Scholar] [PubMed]
- Eich, M.; Roos, W.P.; Nikolova, T.; Kaina, B. Contribution of ATM and ATR to the resistance of glioblastoma and malignant melanoma cells to the methylating anticancer drug temozolomide. Mol. Cancer Ther. 2013, 12, 2529–2540. [Google Scholar] [CrossRef] [PubMed]
- Roos, W.P.; Thomas, A.D.; Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 2016, 16, 20–33. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Roos, W.P.; Wu, Q.; Hofmann, T.G.; Kaina, B. The SIAH1-HIPK2-p53ser46 Damage Response Pathway is Involved in Temozolomide-Induced Glioblastoma Cell Death. Mol. Cancer Res. 2019, 17, 1129–1141. [Google Scholar] [CrossRef] [PubMed]
- Tomicic, M.T.; Meise, R.; Aasland, D.; Berte, N.; Kitzinger, R.; Kramer, O.H.; Kaina, B.; Christmann, M. Apoptosis induced by temozolomide and nimustine in glioblastoma cells is supported by JNK/c-Jun-mediated induction of the BH3-only protein BIM. Oncotarget 2015, 6, 33755–33768. [Google Scholar] [CrossRef] [PubMed]
- Roos, W.P.; Batista, L.F.Z.; Naumann, S.; Wick, W.; Weller, M.; Menck, C.F.M.; Kaina, B. Apoptosis in malignant glioma cells triggered by the temozolomide-induced DNA lesion O6-methylguanine. Oncogene 2007, 26, 186–197. [Google Scholar] [CrossRef]
- Batista, L.F.; Roos, W.P.; Christmann, M.; Menck, C.F.; Kaina, B. Differential sensitivity of malignant glioma cells to methylating and chloroethylating anticancer drugs: p53 determines the switch by regulating xpc, ddb2, and DNA double-strand breaks. Cancer Res. 2007, 67, 11886–11895. [Google Scholar] [CrossRef]
- Knizhnik, A.V.; Roos, W.P.; Nikolova, T.; Quiros, S.; Tomaszowski, K.H.; Christmann, M.; Kaina, B. Survival and death strategies in glioma cells: Autophagy, senescence and apoptosis triggered by a single type of temozolomide-induced DNA damage. PLoS ONE 2013, 8, e55665. [Google Scholar] [CrossRef]
- Beltzig, L.; Christmann, M.; Kaina, B. Abrogation of Cellular Senescence Induced by Temozolomide in Glioblastoma Cells: Search for Senolytics. Cells 2022, 11, 2588. [Google Scholar] [CrossRef]
- Zhang, L.; Pitcher, L.E.; Prahalad, V.; Niedernhofer, L.J.; Robbins, P.D. Targeting cellular senescence with senotherapeutics: Senolytics and senomorphics. FEBS J. 2023, 290, 1362–1383. [Google Scholar] [CrossRef]
- Wang, F.; Xu, L.; Wen, B.; Song, S.; Zhou, Y.; Wu, H.; Wu, Q.; Wang, Y.; Tong, X.; Yan, H. Ultrasound-excited temozolomide sonosensitization induces necroptosis in glioblastoma. Cancer Lett. 2023, 554, 216033. [Google Scholar] [CrossRef] [PubMed]
- Pagano, C.; Navarra, G.; Coppola, L.; Avilia, G.; Pastorino, O.; Della Monica, R.; Buonaiuto, M.; Torelli, G.; Caiazzo, P.; Bifulco, M.; et al. N6-isopentenyladenosine induces cell death through necroptosis in human glioblastoma cells. Cell Death Discov. 2022, 8, 173. [Google Scholar] [CrossRef] [PubMed]
- Kaina, B.; Ziouta, A.; Ochs, K.; Coquerelle, T. Chromosomal instability, reproductive cell death and apoptosis induced by O6-methylguanine in Mex-, Mex+ and methylation-tolerant mismatch repair compromised cells: Facts and models. Mutat. Res. 1997, 381, 227–241. [Google Scholar] [CrossRef] [PubMed]
- Beltzig, L.; Schwarzenbach, C.; Leukel, P.; Frauenknecht, K.B.M.; Sommer, C.; Tancredi, A.; Hegi, M.E.; Christmann, M.; Kaina, B. Senescence Is the Main Trait Induced by Temozolomide in Glioblastoma Cells. Cancers 2022, 14, 2233. [Google Scholar] [CrossRef] [PubMed]
- Aasland, D.; Gotzinger, L.; Hauck, L.; Berte, N.; Meyer, J.; Effenberger, M.; Schneider, S.; Reuber, E.E.; Roos, W.P.; Tomicic, M.T.; et al. Temozolomide Induces Senescence and Repression of DNA Repair Pathways in Glioblastoma Cells via Activation of ATR-CHK1, p21, and NF-kappaB. Cancer Res. 2019, 79, 99–113. [Google Scholar] [CrossRef] [PubMed]
- Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Demaria, M.; O’Leary, M.N.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–176. [Google Scholar] [CrossRef]
- Bujarrabal-Dueso, A.; Sendtner, G.; Meyer, D.H.; Chatzinikolaou, G.; Stratigi, K.; Garinis, G.A.; Schumacher, B. The DREAM complex functions as conserved master regulator of somatic DNA-repair capacities. Nat. Struct. Mol. Biol. 2023, 30, 475–488. [Google Scholar] [CrossRef]
- Lee, S.W.; Kim, H.K.; Lee, N.H.; Yi, H.Y.; Kim, H.S.; Hong, S.H.; Hong, Y.K.; Joe, Y.A. The synergistic effect of combination temozolomide and chloroquine treatment is dependent on autophagy formation and p53 status in glioma cells. Cancer Lett. 2015, 360, 195–204. [Google Scholar] [CrossRef]
- Hori, Y.S.; Hosoda, R.; Akiyama, Y.; Sebori, R.; Wanibuchi, M.; Mikami, T.; Sugino, T.; Suzuki, K.; Maruyama, M.; Tsukamoto, M.; et al. Chloroquine potentiates temozolomide cytotoxicity by inhibiting mitochondrial autophagy in glioma cells. J. Neuro-Oncol. 2015, 122, 11–20. [Google Scholar] [CrossRef] [PubMed]
- Weyerhauser, P.; Kantelhardt, S.R.; Kim, E.L. Re-purposing Chloroquine for Glioblastoma: Potential Merits and Confounding Variables. Front. Oncol. 2018, 8, 335. [Google Scholar] [CrossRef] [PubMed]
- Briceno, E.; Reyes, S.; Sotelo, J. Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine. Neurosurg. Focus 2003, 14, e3. [Google Scholar] [CrossRef] [PubMed]
- Bilger, A.; Bittner, M.I.; Grosu, A.L.; Wiedenmann, N.; Meyer, P.T.; Firat, E.; Niedermann, G.; Weber, W.A.; Milanovic, D. FET-PET-based reirradiation and chloroquine in patients with recurrent glioblastoma: First tolerability and feasibility results. Strahlenther. Onkol. 2014, 190, 957–961. [Google Scholar] [CrossRef] [PubMed]
- Rosenfeld, M.R.; Ye, X.; Supko, J.G.; Desideri, S.; Grossman, S.A.; Brem, S.; Mikkelson, T.; Wang, D.; Chang, Y.C.; Hu, J.; et al. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 2014, 10, 1359–1368. [Google Scholar] [CrossRef] [PubMed]
- Compter, I.; Eekers, D.B.P.; Hoeben, A.; Rouschop, K.M.A.; Reymen, B.; Ackermans, L.; Beckervordersantforth, J.; Bauer, N.J.C.; Anten, M.M.; Wesseling, P.; et al. Chloroquine combined with concurrent radiotherapy and temozolomide for newly diagnosed glioblastoma: A phase IB trial. Autophagy 2021, 17, 2604–2612. [Google Scholar] [CrossRef] [PubMed]
- Ou, M.; Cho, H.Y.; Fu, J.; Thein, T.Z.; Wang, W.; Swenson, S.D.; Minea, R.O.; Stathopoulos, A.; Schonthal, A.H.; Hofman, F.M.; et al. Inhibition of autophagy and induction of glioblastoma cell death by NEO214, a perillyl alcohol-rolipram conjugate. Autophagy 2023, 19, 3169–3188. [Google Scholar] [CrossRef]
- Cho, H.Y.; Wang, W.; Jhaveri, N.; Lee, D.J.; Sharma, N.; Dubeau, L.; Schonthal, A.H.; Hofman, F.M.; Chen, T.C. NEO212, temozolomide conjugated to perillyl alcohol, is a novel drug for effective treatment of a broad range of temozolomide-resistant gliomas. Mol. Cancer Ther. 2014, 13, 2004–2017. [Google Scholar] [CrossRef]
- Strobel, H.; Baisch, T.; Fitzel, R.; Schilberg, K.; Siegelin, M.D.; Karpel-Massler, G.; Debatin, K.M.; Westhoff, M.A. Temozolomide and Other Alkylating Agents in Glioblastoma Therapy. Biomedicines 2019, 7, 69. [Google Scholar] [CrossRef]
- He, Y.; Kaina, B. Are There Thresholds in Glioblastoma Cell Death Responses Triggered by Temozolomide? Int. J. Mol. Sci. 2019, 20, 1562. [Google Scholar] [CrossRef]
- Stratenwerth, B.; Geisen, S.M.; He, Y.; Beltzig, L.; Sturla, S.J.; Kaina, B. Molecular Dosimetry of Temozolomide: Quantification of Critical Lesions, Correlation to Cell Death Responses, and Threshold Doses. Mol. Cancer Ther. 2021, 20, 1789–1799. [Google Scholar] [CrossRef] [PubMed]
- Gilbert, M.R.; Wang, M.; Aldape, K.D.; Stupp, R.; Hegi, M.E.; Jaeckle, K.A.; Armstrong, T.S.; Wefel, J.S.; Won, M.; Blumenthal, D.T.; et al. Dose-dense temozolomide for newly diagnosed glioblastoma: A randomized phase III clinical trial. J. Clin. Oncol. 2013, 31, 4085–4091. [Google Scholar] [CrossRef] [PubMed]
- Christmann, M.; Nagel, G.; Horn, S.; Krahn, U.; Wiewrodt, D.; Sommer, C.; Kaina, B. MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: A comparative study on astrocytoma and glioblastoma. Int. J. Cancer 2010, 127, 2106–2118. [Google Scholar] [CrossRef] [PubMed]
- Sciuscio, D.; Diserens, A.C.; van Dommelen, K.; Martinet, D.; Jones, G.; Janzer, R.C.; Pollo, C.; Hamou, M.F.; Kaina, B.; Stupp, R.; et al. Extent and Patterns of MGMT Promoter Methylation in Glioblastoma- and Respective Glioblastoma-Derived Spheres. Clin. Cancer Res. 2011, 17, 255–266. [Google Scholar] [CrossRef]
- Strik, H.M.; Buhk, J.H.; Wrede, A.; Hoffmann, A.L.; Bock, H.C.; Christmann, M.; Kaina, B. Rechallenge with temozolomide with different scheduling is effective in recurrent malignant gliomas. Mol. Med. Rep. 2008, 1, 863–867. [Google Scholar] [CrossRef]
- Fritz, G.; Tano, K.; Mitra, S.; Kaina, B. Inducibility of the DNA repair gene encoding O6-methylguanine-DNA methyltransferase in mammalian cells by DNA-damaging treatments. Mol. Cell. Biol. 1991, 11, 4660–4668. [Google Scholar]
- Aasland, D.; Reich, T.R.; Tomicic, M.T.; Switzeny, O.J.; Kaina, B.; Christmann, M. Repair gene O(6) -methylguanine-DNA methyltransferase is controlled by SP1 and up-regulated by glucocorticoids, but not by temozolomide and radiation. J. Neurochem. 2018, 144, 139–151. [Google Scholar] [CrossRef]
- Pitter, K.L.; Tamagno, I.; Alikhanyan, K.; Hosni-Ahmed, A.; Pattwell, S.S.; Donnola, S.; Dai, C.; Ozawa, T.; Chang, M.; Chan, T.A.; et al. Corticosteroids compromise survival in glioblastoma. Brain 2016, 139, 1458–1471. [Google Scholar] [CrossRef]
- Tancredi, A.; Gusyatiner, O.; Bady, P.; Buri, M.C.; Lomazzi, R.; Chiesi, D.; Messerer, M.; Hegi, M.E. BET protein inhibition sensitizes glioblastoma cells to temozolomide treatment by attenuating MGMT expression. Cell Death Dis. 2022, 13, 1037. [Google Scholar] [CrossRef]
- Kaina, B.; Margison, G.P.; Christmann, M. Targeting O-methylguanine-DNA methyltransferase with specific inhibitors as a strategy in cancer therapy. Cell Mol. Life Sci. 2010, 67, 3663–3681. [Google Scholar] [CrossRef]
- Jansen, M.; Sorg, U.R.; Ragg, S.; Flasshove, M.; Seeber, S.; Williams, D.A.; Moritz, T. Hematoprotection and enrichment of transduced cells in vivo after gene transfer of MGMT(P140K) into hematopoietic stem cells. Cancer Gene Ther. 2002, 9, 737–746. [Google Scholar] [CrossRef] [PubMed]
- Kaina, B.; Muhlhausen, U.; Piee-Staffa, A.; Christmann, M.; Garcia Boy, R.; Rosch, F.; Schirrmacher, R. Inhibition of O6-methylguanine-DNA methyltransferase by glucose-conjugated inhibitors: Comparison with nonconjugated inhibitors and effect on fotemustine and temozolomide-induced cell death. J. Pharmacol. Exp. Ther. 2004, 311, 585–593. [Google Scholar] [CrossRef]
- Tomaszowski, K.H.; Hellmann, N.; Ponath, V.; Takatsu, H.; Shin, H.W.; Kaina, B. Uptake of glucose-conjugated MGMT inhibitors in cancer cells: Role of flippases and type IV P-type ATPases. Sci. Rep. 2017, 7, 13925. [Google Scholar] [CrossRef] [PubMed]
- Tomaszowski, K.H.; Schirrmacher, R.; Kaina, B. Multidrug Efflux Pumps Attenuate the Effect of MGMT Inhibitors. Mol. Pharm. 2015, 12, 3924–3934. [Google Scholar] [CrossRef] [PubMed]
- Koch, D.; Hundsberger, T.; Boor, S.; Kaina, B. Local intracerebral administration of O(6)-benzylguanine combined with systemic chemotherapy with temozolomide of a patient suffering from a recurrent glioblastoma. J. Neuro-Oncol. 2007, 82, 85–89. [Google Scholar] [CrossRef] [PubMed]
- Vieito, M.; Simonelli, M.; de Vos, F.; Moreno, V.; Geurts, M.; Lorenzi, E.; Macchini, M.; van den Bent, M.J.; Del Conte, G.; de Jonge, M.; et al. Trotabresib (CC-90010) in combination with adjuvant temozolomide or concomitant temozolomide plus radiotherapy in patients with newly diagnosed glioblastoma. Neuro-Oncol. Adv. 2022, 4, vdac146. [Google Scholar] [CrossRef] [PubMed]
- Bale, T.A.; Rosenblum, M.K. The 2021 WHO Classification of Tumors of the Central Nervous System: An update on pediatric low-grade gliomas and glioneuronal tumors. Brain Pathol. 2022, 32, e13060. [Google Scholar] [CrossRef]
- Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Figarella-Branger, D.; Cavenee, W.K.; Ohgaki, H.; Wiestler, O.D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef]
- Zhao, S.; Guan, K.L. IDH1 mutant structures reveal a mechanism of dominant inhibition. Cell Res. 2010, 20, 1279–1281. [Google Scholar] [CrossRef]
- Cohen, A.L.; Holmen, S.L.; Colman, H. IDH1 and IDH2 mutations in gliomas. Curr. Neurol. Neurosci. Rep. 2013, 13, 345. [Google Scholar] [CrossRef]
- Liu, Y.; Hu, H.; Zhang, C.; Wang, Z.; Li, M.; Zhang, W.; Jiang, T. Methylation associated genes contribute to the favorable prognosis of gliomas with isocitrate dehydrogenase 1 mutation. Am. J. Cancer Res. 2015, 5, 2745–2755. [Google Scholar] [PubMed]
- Chowdhury, R.; Yeoh, K.K.; Tian, Y.M.; Hillringhaus, L.; Bagg, E.A.; Rose, N.R.; Leung, I.K.; Li, X.S.; Woon, E.C.; Yang, M.; et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 2011, 12, 463–469. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, C.; Hentschel, B.; Wick, W.; Capper, D.; Felsberg, J.; Simon, M.; Westphal, M.; Schackert, G.; Meyermann, R.; Pietsch, T.; et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: Implications for classification of gliomas. Acta Neuropathol. 2010, 120, 707–718. [Google Scholar] [CrossRef]
- van den Bent, M.J.; Dubbink, H.J.; Marie, Y.; Brandes, A.A.; Taphoorn, M.J.; Wesseling, P.; Frenay, M.; Tijssen, C.C.; Lacombe, D.; Idbaih, A.; et al. IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: A report of the European Organization for Research and Treatment of Cancer Brain Tumor Group. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 1597–1604. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.B.; Dong, D.F.; Wang, M.D.; Gao, K. IDH1 overexpression induced chemotherapy resistance and IDH1 mutation enhanced chemotherapy sensitivity in Glioma cells in vitro and in vivo. Asian Pac. J. Cancer Prev. 2014, 15, 427–432. [Google Scholar] [CrossRef]
- Houillier, C.; Wang, X.; Kaloshi, G.; Mokhtari, K.; Guillevin, R.; Laffaire, J.; Paris, S.; Boisselier, B.; Idbaih, A.; Laigle-Donadey, F.; et al. IDH1 or IDH2 mutations predict longer survival and response to temozolomide in low-grade gliomas. Neurology 2010, 75, 1560–1566. [Google Scholar] [CrossRef]
- Tran, A.N.; Lai, A.; Li, S.; Pope, W.B.; Teixeira, S.; Harris, R.J.; Woodworth, D.C.; Nghiemphu, P.L.; Cloughesy, T.F.; Ellingson, B.M. Increased sensitivity to radiochemotherapy in IDH1 mutant glioblastoma as demonstrated by serial quantitative MR volumetry. Neuro-Oncology 2014, 16, 414–420. [Google Scholar] [CrossRef]
- Li, S.; Chou, A.P.; Chen, W.; Chen, R.; Deng, Y.; Phillips, H.S.; Selfridge, J.; Zurayk, M.; Lou, J.J.; Everson, R.G.; et al. Overexpression of isocitrate dehydrogenase mutant proteins renders glioma cells more sensitive to radiation. Neuro-Oncology 2013, 15, 57–68. [Google Scholar] [CrossRef]
- Ohba, S.; Mukherjee, J.; See, W.L.; Pieper, R.O. Mutant IDH1-driven cellular transformation increases RAD51-mediated homologous recombination and temozolomide resistance. Cancer Res. 2014, 74, 4836–4844. [Google Scholar] [CrossRef]
- Sulkowski, P.L.; Corso, C.D.; Robinson, N.D.; Scanlon, S.E.; Purshouse, K.R.; Bai, H.; Liu, Y.; Sundaram, R.K.; Hegan, D.C.; Fons, N.R.; et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci. Transl. Med. 2017, 9, eaal2463. [Google Scholar] [CrossRef]
- Dubbink, H.J.; Taal, W.; van Marion, R.; Kros, J.M.; van Heuvel, I.; Bromberg, J.E.; Zonnenberg, B.A.; Zonnenberg, C.B.; Postma, T.J.; Gijtenbeek, J.M.; et al. IDH1 mutations in low-grade astrocytomas predict survival but not response to temozolomide. Neurology 2009, 73, 1792–1795. [Google Scholar] [CrossRef] [PubMed]
- Shi, J.; Sun, B.; Shi, W.; Zuo, H.; Cui, D.; Ni, L.; Chen, J. Decreasing GSH and increasing ROS in chemosensitivity gliomas with IDH1 mutation. Tumour Biol. J. Int. Soc. Oncodev. Biol. Med. 2015, 36, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Chaturvedi, A.; Araujo Cruz, M.M.; Jyotsana, N.; Sharma, A.; Yun, H.; Gorlich, K.; Wichmann, M.; Schwarzer, A.; Preller, M.; Thol, F.; et al. Mutant IDH1 promotes leukemogenesis in vivo and can be specifically targeted in human AML. Blood 2013, 122, 2877–2887. [Google Scholar] [CrossRef] [PubMed]
- Mohrenz, I.V.; Antonietti, P.; Pusch, S.; Capper, D.; Balss, J.; Voigt, S.; Weissert, S.; Mukrowsky, A.; Frank, J.; Senft, C.; et al. Isocitrate dehydrogenase 1 mutant R132H sensitizes glioma cells to BCNU-induced oxidative stress and cell death. Apoptosis Int. J. Program. Cell Death 2013, 18, 1416–1425. [Google Scholar] [CrossRef] [PubMed]
- Gilbert, M.R.; Liu, Y.; Neltner, J.; Pu, H.; Morris, A.; Sunkara, M.; Pittman, T.; Kyprianou, N.; Horbinski, C. Autophagy and oxidative stress in gliomas with IDH1 mutations. Acta Neuropathol. 2014, 127, 221–233. [Google Scholar] [CrossRef] [PubMed]
- Mellinghoff, I.K.; Wang, M.Y.; Vivanco, I.; Haas-Kogan, D.A.; Zhu, S.; Dia, E.Q.; Lu, K.V.; Yoshimoto, K.; Huang, J.H.; Chute, D.J.; et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 2005, 353, 2012–2024. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Wu, C.; Chen, N.; Gu, H.; Yen, A.; Cao, L.; Wang, E.; Wang, L. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget 2016, 7, 33440–33450. [Google Scholar] [CrossRef]
- Liu, F.; Hon, G.C.; Villa, G.R.; Turner, K.M.; Ikegami, S.; Yang, H.; Ye, Z.; Li, B.; Kuan, S.; Lee, A.Y.; et al. EGFR Mutation Promotes Glioblastoma through Epigenome and Transcription Factor Network Remodeling. Mol. Cell 2015, 60, 307–318. [Google Scholar] [CrossRef]
- Chiesa, S.; Mangraviti, A.; Martini, M.; Cenci, T.; Mazzarella, C.; Gaudino, S.; Bracci, S.; Martino, A.; Della Pepa, G.M.; Offi, M.; et al. Clinical and NGS predictors of response to regorafenib in recurrent glioblastoma. Sci. Rep. 2022, 12, 16265. [Google Scholar] [CrossRef]
- Chaudhuri, A.R.; Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 610–621. [Google Scholar] [CrossRef]
- Nikolova, T.; Roos, W.P.; Kramer, O.H.; Strik, H.M.; Kaina, B. Chloroethylating nitrosoureas in cancer therapy: DNA damage, repair and cell death signaling. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 29–39. [Google Scholar] [CrossRef] [PubMed]
- Tomicic, M.T.; Aasland, D.; Naumann, S.C.; Meise, R.; Barckhausen, C.; Kaina, B.; Christmann, M. Translesion polymerase eta is upregulated by cancer therapeutics and confers anticancer drug resistance. Cancer Res. 2014, 74, 5585–5596. [Google Scholar] [CrossRef] [PubMed]
- Haracska, L.; Prakash, S.; Prakash, L. Replication past O(6)-methylguanine by yeast and human DNA polymerase eta. Mol. Cell Biol. 2000, 20, 8001–8007. [Google Scholar] [CrossRef] [PubMed]
- Roos, W.P.; Tsaalbi-Shtylik, A.; Tsaryk, R.; Guvercin, F.; de Wind, N.; Kaina, B. The translesion polymerase Rev3L in the tolerance of alkylating anticancer drugs. Mol. Pharmacol. 2009, 76, 927–934. [Google Scholar] [CrossRef] [PubMed]
- Matt, S.; Hofmann, T.G. The DNA damage-induced cell death response: A roadmap to kill cancer cells. Cell Mol. Life Sci. 2016, 73, 2829–2850. [Google Scholar] [CrossRef] [PubMed]
- Tomita, Y.; Marchenko, N.; Erster, S.; Nemajerova, A.; Dehner, A.; Klein, C.; Pan, H.; Kessler, H.; Pancoska, P.; Moll, U.M. WT p53, but not tumor-derived mutants, bind to Bcl2 via the DNA binding domain and induce mitochondrial permeabilization. J. Biol. Chem. 2006, 281, 8600–8606. [Google Scholar] [CrossRef]
- Matissek, K.J.; Mossalam, M.; Okal, A.; Lim, C.S. The DNA binding domain of p53 is sufficient to trigger a potent apoptotic response at the mitochondria. Mol. Pharm. 2013, 10, 3592–3602. [Google Scholar] [CrossRef]
- Kaina, B.; Christmann, M. DNA repair in personalized brain cancer therapy with temozolomide and nitrosoureas. DNA Repair 2019, 78, 128–141. [Google Scholar] [CrossRef]
- Quiros, S.; Roos, W.P.; Kaina, B. Rad51 and BRCA2—New molecular targets for sensitizing glioma cells to alkylating anticancer drugs. PLoS ONE 2011, 6, e27183. [Google Scholar] [CrossRef]
- Sharma, M.; Barravecchia, I.; Teis, R.; Cruz, J.; Mumby, R.; Ziemke, E.K.; Espinoza, C.E.; Krishnamoorthy, V.; Magnuson, B.; Ljungman, M.; et al. Targeting DNA Repair and Survival Signaling in Diffuse Intrinsic Pontine Gliomas to Prevent Tumor Recurrence. Mol. Cancer Ther. 2023. [Google Scholar] [CrossRef]
- Roos, W.P.; Frohnapfel, L.; Quiros, S.; Ringel, F.; Kaina, B. XRCC3 contributes to temozolomide resistance of glioblastoma cells by promoting DNA double-strand break repair. Cancer Lett. 2018, 424, 119–126. [Google Scholar] [CrossRef] [PubMed]
- 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-Oncology 2021, 23, 920–931. [Google Scholar] [CrossRef] [PubMed]
- Cropper, J.D.; Alimbetov, D.S.; Brown, K.T.G.; Likhotvorik, R.I.; Robles, A.J.; Guerra, J.T.; He, B.; Chen, Y.; Kwon, Y.; Kurmasheva, R.T. PARP1-MGMT complex underpins pathway crosstalk in O6-methylguanine repair. J. Hematol. Oncol. 2022, 15, 146. [Google Scholar] [CrossRef] [PubMed]
- Shields, L.B.; Shelton, B.J.; Shearer, A.J.; Chen, L.; Sun, D.A.; Parsons, S.; Bourne, T.D.; LaRocca, R.; Spalding, A.C. Dexamethasone administration during definitive radiation and temozolomide renders a poor prognosis in a retrospective analysis of newly diagnosed glioblastoma patients. Radiat. Oncol. 2015, 10, 222. [Google Scholar] [CrossRef] [PubMed]
- Ueda, S.; Mineta, T.; Nakahara, Y.; Okamoto, H.; Shiraishi, T.; Tabuchi, K. Induction of the DNA repair gene O6-methylguanine-DNA methyltransferase by dexamethasone in glioblastomas. J. Neurosurg. 2004, 101, 659–663. [Google Scholar] [CrossRef]
- Stupp, R.; Hegi, M.E.; Mason, W.P.; van den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef]
- Bobola, M.S.; Kolstoe, D.D.; Blank, A.; Silber, J.R. Minimally cytotoxic doses of temozolomide produce radiosensitization in human glioblastoma cells regardless of MGMT expression. Mol. Cancer Ther. 2010, 9, 1208–1218. [Google Scholar] [CrossRef]
- Kaina, B.; Beltzig, L.; Strik, H. Temozolomide—Just a Radiosensitizer? Front. Oncol. 2022, 12, 912821. [Google Scholar] [CrossRef]
- Clark, P.A.; Gaal, J.T.; Strebe, J.K.; Pasch, C.A.; Deming, D.A.; Kuo, J.S.; Robins, H.I. The effects of tumor treating fields and temozolomide in MGMT expressing and non-expressing patient-derived glioblastoma cells. J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas. 2017, 36, 120–124. [Google Scholar] [CrossRef]
- Stupp, R.; Taillibert, S.; Kanner, A.A.; Kesari, S.; Steinberg, D.M.; Toms, S.A.; Taylor, L.P.; Lieberman, F.; Silvani, A.; Fink, K.L.; et al. Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma: A Randomized Clinical Trial. JAMA 2015, 314, 2535–2543. [Google Scholar] [CrossRef]
- Giladi, M.; Schneiderman, R.S.; Voloshin, T.; Porat, Y.; Munster, M.; Blat, R.; Sherbo, S.; Bomzon, Z.; Urman, N.; Itzhaki, A.; et al. Mitotic Spindle Disruption by Alternating Electric Fields Leads to Improper Chromosome Segregation and Mitotic Catastrophe in Cancer Cells. Sci. Rep. 2015, 5, 18046. [Google Scholar] [CrossRef] [PubMed]
- Giladi, M.; Munster, M.; Schneiderman, R.S.; Voloshin, T.; Porat, Y.; Blat, R.; Zielinska-Chomej, K.; Haag, P.; Bomzon, Z.; Kirson, E.D.; et al. Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cells. Radiat. Oncol. 2017, 12, 206. [Google Scholar] [CrossRef] [PubMed]
- Guo, X.; Yang, X.; Wu, J.; Yang, H.; Li, Y.; Li, J.; Liu, Q.; Wu, C.; Xing, H.; Liu, P.; et al. Tumor-Treating Fields in Glioblastomas: Past, Present, and Future. Cancers 2022, 14, 3669. [Google Scholar] [CrossRef] [PubMed]
- Fishman, H.; Monin, R.; Dor-On, E.; Kinzel, A.; Haber, A.; Giladi, M.; Weinberg, U.; Palti, Y. Tumor Treating Fields (TTFields) increase the effectiveness of temozolomide and lomustine in glioblastoma cell lines. J. Neuro-Oncol. 2023, 163, 83–94. [Google Scholar] [CrossRef] [PubMed]
- Lazaridis, L.; Bumes, E.; Cacilia Spille, D.; Schulz, T.; Heider, S.; Agkatsev, S.; Schmidt, T.; Blau, T.; Oster, C.; Feldheim, J.; et al. First multicentric real-life experience with the combination of CCNU and temozolomide in newly diagnosed MGMT promoter methylated IDH wildtype glioblastoma. Neuro-Oncol. Adv. 2022, 4, vdac137. [Google Scholar] [CrossRef] [PubMed]
- Kast, R.E.; Karpel-Massler, G.; Halatsch, M.E. CUSP9* treatment protocol for recurrent glioblastoma: Aprepitant, artesunate, auranofin, captopril, celecoxib, disulfiram, itraconazole, ritonavir, sertraline augmenting continuous low dose temozolomide. Oncotarget 2014, 5, 8052–8082. [Google Scholar] [CrossRef]
- Halatsch, M.E.; Kast, R.E.; Karpel-Massler, G.; Mayer, B.; Zolk, O.; Schmitz, B.; Scheuerle, A.; Maier, L.; Bullinger, L.; Mayer-Steinacker, R.; et al. A phase Ib/IIa trial of 9 repurposed drugs combined with temozolomide for the treatment of recurrent glioblastoma: CUSP9v3. Neuro-Oncol. Adv. 2021, 3, vdab075. [Google Scholar] [CrossRef]
- Arrieta, V.A.; Dmello, C.; McGrail, D.J.; Brat, D.J.; Lee-Chang, C.; Heimberger, A.B.; Chand, D.; Stupp, R.; Sonabend, A.M. Immune checkpoint blockade in glioblastoma: From tumor heterogeneity to personalized treatment. J. Clin. Investig. 2023, 133, e163447. [Google Scholar] [CrossRef]
- Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro-Oncology 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
- Daniel, P.; Meehan, B.; Sabri, S.; Jamali, F.; Sarkaria, J.N.; Choi, D.; Garnier, D.; Kitange, G.; Glennon, K.I.; Paccard, A.; et al. Detection of temozolomide-induced hypermutation and response to PD-1 checkpoint inhibitor in recurrent glioblastoma. Neuro-Oncol. Adv. 2022, 4, vdac076. [Google Scholar] [CrossRef]
- Franzese, O.; Torino, F.; Giannetti, E.; Cioccoloni, G.; Aquino, A.; Faraoni, I.; Fuggetta, M.P.; De Vecchis, L.; Giuliani, A.; Kaina, B.; et al. Abscopal Effect and Drug-Induced Xenogenization: A Strategic Alliance in Cancer Treatment? Int. J. Mol. Sci. 2021, 22, 10672. [Google Scholar] [CrossRef]
- Juric, V.; Murphy, B. Cyclin-dependent kinase inhibitors in brain cancer: Current state and future directions. Cancer Drug Resist. 2020, 3, 48–62. [Google Scholar] [CrossRef] [PubMed]
- Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Ruda, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib compared with lomustine in patients with relapsed glioblastoma (REGOMA): A multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef] [PubMed]
- Ruda, R.; Bruno, F.; Pellerino, A.; Pronello, E.; Palmiero, R.; Bertero, L.; Crasto, S.; Polo, V.; Vitaliani, R.; Trincia, E.; et al. Observational real-life study on regorafenib in recurrent glioblastoma: Does dose reduction reduce toxicity while maintaining the efficacy? J. Neuro-Oncol. 2022, 160, 389–402. [Google Scholar] [CrossRef]
- Mongiardi, M.P.; Buccarelli, M.; Formato, A.; Orecchini, E.; Salbini, M.; Ricci, V.; Orsini, T.; Putti, S.; Chiesa, S.; Ricci-Vitiani, L.; et al. Characterization of Glioblastoma Cells Response to Regorafenib. Cancers 2022, 14, 6193. [Google Scholar] [CrossRef] [PubMed]
- Roos, W.; Baumgartner, M.; Kaina, B. Apoptosis triggered by DNA damage O6-methylguanine in human lymphocytes requires DNA replication and is mediated by p53 and Fas/CD95/Apo-1. Oncogene 2004, 23, 359–367. [Google Scholar] [CrossRef]
- Wong, E.; Nahar, N.; Hau, E.; Varikatt, W.; Gebski, V.; Ng, T.; Jayamohan, J.; Sundaresan, P. Cut-point for Ki-67 proliferation index as a prognostic marker for glioblastoma. Asia-Pac. J. Clin. Oncol. 2019, 15, 5–9. [Google Scholar] [CrossRef]
- Bredel, M.; Piribauer, M.; Marosi, C.; Birner, P.; Gatterbauer, B.; Fischer, I.; Strobel, T.; Rossler, K.; Budka, H.; Hainfellner, J.A. High expression of DNA topoisomerase IIalpha and Ki-67 antigen is associated with prolonged survival in glioblastoma patients. Eur. J. Cancer 2002, 38, 1343–1347. [Google Scholar] [CrossRef]
- Beltzig, L.; Stratenwerth, B.; Kaina, B. Accumulation of Temozolomide-Induced Apoptosis, Senescence and DNA Damage by Metronomic Dose Schedule: A Proof-of-Principle Study with Glioblastoma Cells. Cancers 2021, 13, 6287. [Google Scholar] [CrossRef]
- Fu, D.; Calvo, J.A.; Samson, L.D. Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat. Rev. Cancer 2012, 12, 104–120. [Google Scholar] [CrossRef]
- Ensminger, M.; Iloff, L.; Ebel, C.; Nikolova, T.; Kaina, B.; Lbrich, M. DNA breaks and chromosomal aberrations arise when replication meets base excision repair. J. Cell Biol. 2014, 206, 29–43. [Google Scholar] [CrossRef] [PubMed]
- Tang, J.B.; Svilar, D.; Trivedi, R.N.; Wang, X.H.; Goellner, E.M.; Moore, B.; Hamilton, R.L.; Banze, L.A.; Brown, A.R.; Sobol, R.W. N-methylpurine DNA glycosylase and DNA polymerase beta modulate BER inhibitor potentiation of glioma cells to temozolomide. Neuro-Oncology 2011, 13, 471–486. [Google Scholar] [CrossRef] [PubMed]
- Agnihotri, S.; Gajadhar, A.S.; Ternamian, C.; Gorlia, T.; Diefes, K.L.; Mischel, P.S.; Kelly, J.; McGown, G.; Thorncroft, M.; Carlson, B.L.; et al. Alkylpurine-DNA-N-glycosylase confers resistance to temozolomide in xenograft models of glioblastoma multiforme and is associated with poor survival in patients. J. Clin. Investig. 2012, 122, 253–266. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.T.; Rajakannu, P.; Pham, M.D.T.; Weman, L.; Jucht, A.; Buri, M.C.; Van Dommelen, K.; Hegi, M.E. Epigenetic silencing of HTATIP2 in glioblastoma contributes to treatment resistance by enhancing nuclear translocation of the DNA repair protein MPG. Mol. Oncol. 2023, 17, 1744–1762. [Google Scholar] [CrossRef] [PubMed]
- Song, Y.Q.; Li, G.D.; Niu, D.; Chen, F.; Jing, S.; Wai Wong, V.K.; Wang, W.; Leung, C.H. A robust luminescent assay for screening alkyladenine DNA glycosylase inhibitors to overcome DNA repair and temozolomide drug resistance. J. Pharm. Anal. 2023, 13, 514–522. [Google Scholar] [CrossRef] [PubMed]
- Cahill, D.P.; Levine, K.K.; Betensky, R.A.; Codd, P.J.; Romany, C.A.; Reavie, L.B.; Batchelor, T.T.; Futreal, P.A.; Stratton, M.R.; Curry, W.T.; et al. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin. Cancer Res. 2007, 13, 2038–2045. [Google Scholar] [CrossRef] [PubMed]
- Dosch, J.; Christmann, M.; Kaina, B. Mismatch G-T binding activity and MSH2 expression is quantitatively related to sensitivity of cells to methylating agents. Carcinogenesis 1998, 4, 567–573. [Google Scholar] [CrossRef] [PubMed]
- Maxwell, J.A.; Johnson, S.P.; McLendon, R.E.; Lister, D.W.; Horne, K.S.; Rasheed, A.; Quinn, J.A.; Ali-Osman, F.; Friedman, A.H.; Modrich, P.L.; et al. Mismatch repair deficiency does not mediate clinical resistance to temozolomide in malignant glioma. Clin. Cancer Res. 2008, 14, 4859–4868. [Google Scholar] [CrossRef]
- McFaline-Figueroa, J.L.; Braun, C.J.; Stanciu, M.; Nagel, Z.D.; Mazzucato, P.; Sangaraju, D.; Cerniauskas, E.; Barford, K.; Vargas, A.; Chen, Y.; et al. Minor Changes in Expression of the Mismatch Repair Protein MSH2 Exert a Major Impact on Glioblastoma Response to Temozolomide. Cancer Res. 2015, 75, 3127–3138. [Google Scholar] [CrossRef]
- Touat, M.; Li, Y.Y.; Boynton, A.N.; Spurr, L.F.; Iorgulescu, J.B.; Bohrson, C.L.; Cortes-Ciriano, I.; Birzu, C.; Geduldig, J.E.; Pelton, K.; et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature 2020, 580, 517–523. [Google Scholar] [CrossRef]
- Berte, N.; Piee-Staffa, A.; Piecha, N.; Wang, M.; Borgmann, K.; Kaina, B.; Nikolova, T. Targeting Homologous Recombination by Pharmacological Inhibitors Enhances the Killing Response of Glioblastoma Cells Treated with Alkylating Drugs. Mol. Cancer Ther. 2016, 15, 2665–2678. [Google Scholar] [CrossRef]
- Hanisch, D.; Krumm, A.; Diehl, T.; Stork, C.M.; Dejung, M.; Butter, F.; Kim, E.; Brenner, W.; Fritz, G.; Hofmann, T.G.; et al. Class I HDAC overexpression promotes temozolomide resistance in glioma cells by regulating RAD18 expression. Cell Death Dis. 2022, 13, 293. [Google Scholar] [CrossRef]
- Herrlinger, U.; Rieger, J.; Koch, D.; Loeser, S.; Blaschke, B.; Kortmann, R.D.; Steinbach, J.P.; Hundsberger, T.; Wick, W.; Meyermann, R.; et al. Phase II trial of lomustine plus temozolomide chemotherapy in addition to radiotherapy in newly diagnosed glioblastoma: UKT-03. J. Clin. Oncol. 2006, 24, 4412–4417. [Google Scholar] [CrossRef]
- Larsen, B.D.; Sørensen, C.S. The caspase-activated DNase: Apoptosis and beyond. FEBS J. 2017, 284, 1160–1170. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Osuna, M.; Martinez-Escardo, L.; Granados-Colomina, C.; Martinez-Soler, F.; Pascual-Guiral, S.; Iglesias-Guimarais, V.; Velasco, R.; Plans, G.; Vidal, N.; Tortosa, A.; et al. An intrinsic DFF40/CAD endonuclease deficiency impairs oligonucleosomal DNA hydrolysis during caspase-dependent cell death: A common trait in human glioblastoma cells. Neuro-Oncology 2016, 18, 950–961. [Google Scholar] [CrossRef] [PubMed]
- Saleh, T.; Tyutyunyk-Massey, L.; Murray, G.F.; Alotaibi, M.R.; Kawale, A.S.; Elsayed, Z.; Henderson, S.C.; Yakovlev, V.; Elmore, L.W.; Toor, A.; et al. Tumor cell escape from therapy-induced senescence. Biochem. Pharmacol. 2019, 162, 202–212. [Google Scholar] [CrossRef] [PubMed]
- Roberson, R.S.; Kussick, S.J.; Vallieres, E.; Chen, S.Y.; Wu, D.Y. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005, 65, 2795–2803. [Google Scholar] [CrossRef] [PubMed]
- Chojak, R.; Fares, J.; Petrosyan, E.; Lesniak, M.S. Cellular senescence in glioma. J. Neuro-Oncol. 2023, 164, 11–29. [Google Scholar] [CrossRef] [PubMed]
- Schwarzenbach, C.; Tatsch, L.; Brandstetter Vilar, J.; Rasenberger, B.; Beltzig, L.; Kaina, B.; Tomicic, M.T.; Christmann, M. Targeting c-IAP1, c-IAP2, and Bcl-2 Eliminates Senescent Glioblastoma Cells Following Temozolomide Treatment. Cancers 2021, 13, 3585. [Google Scholar] [CrossRef] [PubMed]
- Efferth, T.; Li, P.C.; Konkimalla, V.S.; Kaina, B. From traditional Chinese medicine to rational cancer therapy. Trends Mol. Med. 2007, 13, 353–361. [Google Scholar] [CrossRef]
- Berdelle, N.; Nikolova, T.; Quiros, S.; Efferth, T.; Kaina, B. Artesunate induces oxidative DNA damage, sustained DNA double-strand breaks, and the ATM/ATR damage response in cancer cells. Mol. Cancer Ther. 2011, 10, 2224–2233. [Google Scholar] [CrossRef] [PubMed]
- Li, P.C.; Lam, E.; Roos, W.P.; Zdzienicka, M.Z.; Kaina, B.; Efferth, T. Artesunate derived from traditional Chinese medicine induces DNA damage and repair. Cancer Res. 2008, 68, 4347–4351. [Google Scholar] [CrossRef] [PubMed]
- Efferth, T.; Benakis, A.; Romero, M.R.; Tomicic, M.; Rauh, R.; Steinbach, D.; Hafer, R.; Stamminger, T.; Oesch, F.; Kaina, B.; et al. Enhancement of cytotoxicity of artemisinins toward cancer cells by ferrous iron. Free Radic. Biol. Med. 2004, 37, 998–1009. [Google Scholar] [CrossRef] [PubMed]
- Berte, N.; Lokan, S.; Eich, M.; Kim, E.; Kaina, B. Artesunate enhances the therapeutic response of glioma cells to temozolomide by inhibition of homologous recombination and senescence. Oncotarget 2016, 7, 67235. [Google Scholar] [CrossRef] [PubMed]
- Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. EBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Doornebal, E.J.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D.; Tchkonia, T.; Kirkland, J.L. New agents that target senescent cells: The flavone, fisetin, and the BCL-X(L) inhibitors, A1331852 and A1155463. Aging 2017, 9, 955–963. [Google Scholar] [CrossRef]
- Zhu, Y.; Tchkonia, T.; Pirtskhalava, T.; Gower, A.C.; Ding, H.; Giorgadze, N.; Palmer, A.K.; Ikeno, Y.; Hubbard, G.B.; Lenburg, M.; et al. The Achilles’ heel of senescent cells: From transcriptome to senolytic drugs. Aging Cell 2015, 14, 644–658. [Google Scholar] [CrossRef]
- Barbagallo, G.M.; Paratore, S.; Caltabiano, R.; Palmucci, S.; Parra, H.S.; Privitera, G.; Motta, F.; Lanzafame, S.; Scaglione, G.; Longo, A.; et al. Long-term therapy with temozolomide is a feasible option for newly diagnosed glioblastoma: A single-institution experience with as many as 101 temozolomide cycles. Neurosurg. Focus 2014, 37, E4. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaina, B. Temozolomide, Procarbazine and Nitrosoureas in the Therapy of Malignant Gliomas: Update of Mechanisms, Drug Resistance and Therapeutic Implications. J. Clin. Med. 2023, 12, 7442. https://doi.org/10.3390/jcm12237442
Kaina B. Temozolomide, Procarbazine and Nitrosoureas in the Therapy of Malignant Gliomas: Update of Mechanisms, Drug Resistance and Therapeutic Implications. Journal of Clinical Medicine. 2023; 12(23):7442. https://doi.org/10.3390/jcm12237442
Chicago/Turabian StyleKaina, Bernd. 2023. "Temozolomide, Procarbazine and Nitrosoureas in the Therapy of Malignant Gliomas: Update of Mechanisms, Drug Resistance and Therapeutic Implications" Journal of Clinical Medicine 12, no. 23: 7442. https://doi.org/10.3390/jcm12237442
APA StyleKaina, B. (2023). Temozolomide, Procarbazine and Nitrosoureas in the Therapy of Malignant Gliomas: Update of Mechanisms, Drug Resistance and Therapeutic Implications. Journal of Clinical Medicine, 12(23), 7442. https://doi.org/10.3390/jcm12237442