Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-06-01T15:00:55.553Z Has data issue: false hasContentIssue false

New perspective on DNA response pathway (DDR) in glioblastoma, focus on classic biomarkers and emerging roles of ncRNAs

Published online by Cambridge University Press:  08 May 2023

Bianca Oana Pirlog
Affiliation:
Department of Neuroscience, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
Silvina Ilut
Affiliation:
Department of Neuroscience, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
Radu Pirlog*
Affiliation:
Research Center for Functional Genomics, Biomedicine and Translational Medicine, The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
Paul Chiroi
Affiliation:
Faculty of Medicine, University of Medicine and Pharmacology “Iuliu Hațieganu”, Cluj-Napoca, Romania
Andreea Nutu
Affiliation:
Faculty of Medicine, University of Medicine and Pharmacology “Iuliu Hațieganu”, Cluj-Napoca, Romania
Delia Ioana Radutiu
Affiliation:
Faculty of Medicine, University of Medicine and Pharmacology “Iuliu Hațieganu”, Cluj-Napoca, Romania
George Daniel Cuc
Affiliation:
Research Center for Functional Genomics, Biomedicine and Translational Medicine, The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
Ioana Berindan-Neagoe*
Affiliation:
Faculty of Medicine, University of Medicine and Pharmacology “Iuliu Hațieganu”, Cluj-Napoca, Romania
Seyed Fazel Nabavi
Affiliation:
Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre, Via Cortenocera, 82030, San Salvatore Telesino, BN, Italy Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
Rosanna Filosa
Affiliation:
Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre, Via Cortenocera, 82030, San Salvatore Telesino, BN, Italy Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
Seyed Mohammad Nabavi
Affiliation:
Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre, Via Cortenocera, 82030, San Salvatore Telesino, BN, Italy Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
*
Corresponding authors: Radu Pirlog, Ioana Berindan-Neagoe, Seyed Mohamad Nabavi; Email: pirlog.radu@umfcluj.ro, ioana.neagoe@umfcluj.ro, nabavi208@gmail.com
Corresponding authors: Radu Pirlog, Ioana Berindan-Neagoe, Seyed Mohamad Nabavi; Email: pirlog.radu@umfcluj.ro, ioana.neagoe@umfcluj.ro, nabavi208@gmail.com

Abstract

Background

Glioblastoma (GBM) is the most frequent type of primary brain cancer, having a median survival of only 15 months. The current standard of care includes a combination of surgery, radiotherapy (RT) and chemotherapy with temozolomide, but with limited results. Moreover, multiple studies have shown that tumour relapse and resistance to classic therapeutic approaches are common events that occur in the majority of patients, and eventually leading to death. New approaches to better understand the intricated tumour biology involved in GBM are needed in order to develop personalised treatment approaches. Advances in cancer biology have widen our understanding over the GBM genome and allowing a better classification of these tumours based on their molecular profile.

Methods

A new targeted therapeutic approach that is currently investigated in multiple clinical trials in GBM is represented by molecules that target various defects in the DNA damage repair (DDR) pathway, a mechanism activated by endogenous and exogenous factors that induce alteration of DNA, and is involved for the development of chemotherapy and RT resistance. This intricate pathway is regulated by p53, two important kinases ATR and ATM and non-coding RNAs including microRNAs, long-non-coding RNAs and circular RNAs that regulate the expression of all the proteins involved in the pathway.

Results

Currently, the most studied DDR inhibitors are represented by PARP inhibitors (PARPi) with important results in ovarian and breast cancer. PARPi are a class of tumour agnostic drugs that showed their efficacy also in other localisations such as colon and prostate tumours that have a molecular signature associated with genomic instability. These inhibitors induce the accumulation of intracellular DNA damage, cell cycle arrest, mitotic catastrophe and apoptosis.

Conclusions

This study aims to provide an integrated image of the DDR pathway in glioblastoma under physiological and treatment pressure with a focus of the regulatory roles of ncRNAs. The DDR inhibitors are emerging as an important new therapeutic approach for tumours with genomic instability and alterations in DDR pathways. The first clinical trials with PARPi in GBM are currently ongoing and will be presented in the article. Moreover, we consider that by incorporating the regulatory network in the DDR pathway in GBM we can fill the missing gaps that limited previous attempts to effectively target it in brain tumours. An overview of the importance of ncRNAs in GBM and DDR physiology and how they are interconnected is presented.

Type
Review
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Footnotes

*

These authors contributed equally to this work.

References

Grochans, S et al. (2022) Epidemiology of glioblastoma multiforme–literature review. Cancers 14, 2412.Google ScholarPubMed
Poon, MTC et al. (2020) Longer-term (≥2 years) survival in patients with glioblastoma in population-based studies pre- and post-2005: a systematic review and meta-analysis. Scientific Reports 10, 11622.Google ScholarPubMed
Grech, N et al. (2020) Rising incidence of glioblastoma multiforme in a well-defined population. Cureus 12, e8195.Google Scholar
Tamimi, AF and Juweid, M (2017) Epidemiology and outcome of glioblastoma. In De Vleeschouwer, S (ed.), Glioblastoma [Internet]. Brisbane, AU: Codon Publications, pp. 143153, http://www.ncbi.nlm.nih.gov/books/NBK470003/.Google ScholarPubMed
Louis, DN et al. (2021) The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro-Oncology 23, 12311251.Google ScholarPubMed
Mardis, ER (2019) The impact of next-generation sequencing on cancer genomics: from discovery to clinic. Cold Spring Harbor Perspectives in Medicine 9, a036269.Google ScholarPubMed
Wang, WT et al. (2019) Noncoding RNAs in cancer therapy resistance and targeted drug development. Journal of Hematology Oncology 12, 55.Google ScholarPubMed
Shahzad, U et al. (2021) Noncoding RNAs in glioblastoma: emerging biological concepts and potential therapeutic implications. Cancers 13, 1555.Google ScholarPubMed
Rominiyi, O and Collis, SJ (2022) DDRUgging glioblastoma: understanding and targeting the DNA damage response to improve future therapies. Molecular Oncology 16, 1141.Google ScholarPubMed
Fernandes, C et al. (2017) Current standards of care in glioblastoma therapy. In De Vleeschouwer, S (ed.), Glioblastoma. Brisbane, AU: Codon Publications, pp. 197241, http://www.ncbi.nlm.nih.gov/books/NBK469987/.Google ScholarPubMed
Weller, M et al. (2013) Standards of care for treatment of recurrent glioblastoma--are we there yet? Neuro-Oncology 15, 427.Google ScholarPubMed
Ferri, A et al. (2020) Targeting the DNA damage response to overcome cancer drug resistance in glioblastoma. International Journal of Molecular Sciences 21, 4910.Google ScholarPubMed
Jackson, SP and Bartek, J (2009) The DNA-damage response in human biology and disease. Nature 461, 10711078.Google ScholarPubMed
Everix, L et al. (2022) Perspective on the use of DNA repair inhibitors as a tool for imaging and radionuclide therapy of glioblastoma. Cancers 14, 1821.Google ScholarPubMed
Kaur, E et al. (2022) Glioblastoma recurrent cells switch between ATM and ATR pathway as an alternative strategy to survive radiation stress. Medical Oncology 39, 50.Google ScholarPubMed
Bonm, A and Kesari, S (2021) DNA damage response in glioblastoma: mechanism for treatment resistance and emerging therapeutic strategies. Cancer Journal 27, 379385.Google ScholarPubMed
Caldecott, KW (2008) Single-strand break repair and genetic disease. Nature Reviews Genetics 9, 619631.Google ScholarPubMed
Malyuchenko, NV et al. (2015) PARP1 inhibitors: antitumor drug design. Acta Naturae 7, 2737.Google ScholarPubMed
Leonetti, C et al. (2012) Targeted therapy for brain tumours: role of PARP inhibitors. Current Cancer Drug Targets 12, 218236.Google ScholarPubMed
Sakthikumar, S et al. (2020) Whole-genome sequencing of glioblastoma reveals enrichment of non-coding constraint mutations in known and novel genes. Genome Biology 21, 127.Google ScholarPubMed
Cardon, T et al. (2021) Unveiling a ghost proteome in the glioblastoma non-coding RNAs. Frontiers in Cell and Developmental Biology 9, 703583.Google ScholarPubMed
Zhang, Y et al. (2017) Noncoding RNAs in glioblastoma. In De Vleeschouwer, S (ed.), Glioblastoma. Brisbane, AU: Codon Publications, pp. 95130, http://www.ncbi.nlm.nih.gov/books/NBK469994/.Google ScholarPubMed
Zeng, Z et al. (2022) NcRNAs: multi-angle participation in the regulation of glioma chemotherapy resistance (review). International Journal of Oncology 60, 76.Google ScholarPubMed
Wan, G et al. (2014) Noncoding RNAs in DNA repair and genome integrity. Antioxidants & Redox Signaling 20, 655677.Google ScholarPubMed
Ahmed, SP et al. (2021) Glioblastoma and MiRNAs. Cancers 13, 1581.Google ScholarPubMed
Stackhouse, CT et al. (2020) Exploring the roles of lncRNAs in GBM pathophysiology and their therapeutic potential. Cells 9, E2369.Google ScholarPubMed
Parashar, TR, Ravindran, F and Choudhary, B (2021) DNA Damage Repair Genes and Noncoding RNA in High-Grade Gliomas and Its Clinical Relevance [Internet]. Central Nervous System Tumors. IntechOpen; [cited 2022 Jul 12]. Available at https://www.intechopen.com/chapters/undefined/state.item.id.Google Scholar
Mousavi, SM et al. (2022) Non-coding RNAs and glioblastoma: insight into their roles in metastasis. Molecular Therapy Oncolytics 24, 262287.Google ScholarPubMed
Huang, T et al. (2019) MIR93 (microRNA −93) regulates tumorigenicity and therapy response of glioblastoma by targeting autophagy. Autophagy 15, 11001111.Google ScholarPubMed
DNA Damage Response (DDR) Consortium [Internet]. National Brain Tumor Society. [cited 2022 Jul 24]. Available at https://braintumor.org/our-research/ddr-consortium/.Google Scholar
Weber, AM and Ryan, AJ (2015) ATM and ATR as therapeutic targets in cancer. Pharmacology Therapeutics 149, 124138.Google ScholarPubMed
Repair of Endogenous DNA Damage. [cited 2022 Jul 10]. Available at http://symposium.cshlp.org/content/65/127.extract.Google Scholar
Ciccia, A and Elledge, SJ (2010) The DNA damage response: making it safe to play with knives. Molecular Cell 40, 179204.Google ScholarPubMed
Smith, HL et al. (2020) DNA Damage checkpoint kinases in cancer. Expert Reviews in Molecular Medicine 22, e2.Google ScholarPubMed
Woods, D and Turchi, JJ (2013) Chemotherapy induced DNA damage response. Cancer Biology and Therapy 14, 379389.Google ScholarPubMed
Kaina, B and Christmann, M (2019) DNA repair in personalized brain cancer therapy with temozolomide and nitrosoureas. DNA Repair 78, 128141.Google ScholarPubMed
Ou, A et al. (2020) Molecular mechanisms of treatment resistance in glioblastoma. International Journal of Molecular Sciences 22, E351.Google ScholarPubMed
Aldea, MD et al. (2014) Metformin plus sorafenib highly impacts temozolomide resistant glioblastoma stem-like cells. Journal of the Balkan Union of Oncology 19, 502511.Google ScholarPubMed
Estiar, MA and Mehdipour, P (2018) ATM in breast and brain tumors: a comprehensive review. Cancer Biology & Medicine 15, 210227.Google ScholarPubMed
Zannini, L et al. (2014) CHK2 kinase in the DNA damage response and beyond. Journal of Molecular Cell Biology 6, 442457.Google ScholarPubMed
van Jaarsveld, MTM et al. (2020) Cell-type-specific role of CHK2 in mediating DNA damage-induced G2 cell cycle arrest. Oncogenesis 9, 17.CrossRefGoogle ScholarPubMed
Shen, T and Huang, S (2012) The role of Cdc25A in the regulation of cell proliferation and apoptosis. Anti-Cancer Agents in Medicinal Chemistry 12, 631639.Google ScholarPubMed
Falck, J et al. (2001) The ATM–Chk2–Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 410, 842847.Google ScholarPubMed
Jeggo, PA et al. (2016) DNA repair, genome stability and cancer: a historical perspective. Nature Reviews Cancer 16, 3542.Google Scholar
Fridman, JS and Lowe, SW (2003) Control of apoptosis by p53. Oncogene 22, 90309040.Google ScholarPubMed
Abbas, T and Dutta, A (2009) p21 in cancer: intricate networks and multiple activities. Nature Reviews Cancer 9, 400414.Google ScholarPubMed
Lee, YJ et al. (2020) Gene expression profiling of glioblastoma cell lines depending on TP53 status after tumor-treating fields (TTFields) treatment. Scientific Reports 10, 12272.Google ScholarPubMed
Nam, EA and Cortez, D (2011) ATR signaling: more than meeting at the fork. Biochemical Journal 436, 527536.Google ScholarPubMed
Sirbu, BM and Cortez, D (2013) DNA damage response: three levels of DNA repair regulation. Cold Spring Harbor Perspectives in Biology 5, a012724.Google ScholarPubMed
Rao, Q et al. (2018) Cryo-EM structure of human ATR-ATRIP complex. Cell Research 28, 143156.Google ScholarPubMed
Saldivar, JC et al. (2017) The essential kinase ATR: ensuring faithful duplication of a challenging genome. Nature Reviews Molecular Cell Biology 18, 622636.Google ScholarPubMed
Guo, C et al. (2015) Interaction of Chk1 with Treslin negatively regulates the initiation of chromosomal DNA replication. Molecular Cell 57, 492505.Google ScholarPubMed
Esposito, F et al. (2021) Wee1 kinase: a potential target to overcome tumor resistance to therapy. International Journal of Molecular Sciences 22, 10689.Google ScholarPubMed
Strobel, H et al. (2019) Temozolomide and other alkylating agents in glioblastoma therapy. Biomedicines 7, 69.Google ScholarPubMed
Lee, SY (2016) Temozolomide resistance in glioblastoma multiforme. Genes & Diseases 3, 198210.Google ScholarPubMed
Cui, B et al. (2010) Decoupling of DNA damage response signaling from DNA damages underlies temozolomide resistance in glioblastoma cells. The Journal of Biomedical Research 24, 424435.Google ScholarPubMed
Singh, N et al. (2021) Mechanisms of temozolomide resistance in glioblastoma - a comprehensive review. Cancer Drug Resistance 4, 1743.Google ScholarPubMed
Kitange, GJ et al. (2009) Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts. Neuro-Oncology 11, 281291.Google ScholarPubMed
Chien, CH et al. (2021) Dissecting the mechanism of temozolomide resistance and its association with the regulatory roles of intracellular reactive oxygen species in glioblastoma. Journal of Biomedical Sciences 28, 18.Google ScholarPubMed
Zhang, X et al. (2022) Acquired temozolomide resistance in MGMTlow gliomas is associated with regulation of homologous recombination repair by ROCK2. Cell Death Disease 13, 115.Google ScholarPubMed
Touat, M et al. (2020) Mechanisms and therapeutic implications of hypermutation in gliomas. Nature 580, 517523.Google ScholarPubMed
Bateman, AC (2021) DNA mismatch repair proteins: scientific update and practical guide. Journal of Clinical Pathology 74, 264268.Google ScholarPubMed
McCarthy, AJ et al. (2018) Heterogenous loss of mismatch repair (MMR) protein expression: a challenge for immunohistochemical interpretation and microsatellite instability (MSI) evaluation. The Journal of Pathology: Clinical Research 5, 115129.Google ScholarPubMed
Borrego-Soto, G et al. (2015) Ionizing radiation-induced DNA injury and damage detection in patients with breast cancer. Genetics and Molecular Biology 38, 420432.Google ScholarPubMed
Gzell, C et al. (2017) Radiotherapy in glioblastoma: the past, the present and the future. Clinical Oncology 29, 1525.Google ScholarPubMed
Ray Chaudhuri, A and Nussenzweig, A (2017) The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nature Reviews Molecular Cellular Biology 18, 610621.Google ScholarPubMed
Carruthers, RD et al. (2018) Replication stress drives constitutive activation of the DNA damage response and radioresistance in glioblastoma stem-like cells. Cancer Research 78, 50605071.Google ScholarPubMed
Krajewska, M et al. (2015) Regulators of homologous recombination repair as novel targets for cancer treatment. Frontiers Genetic 6, 96.Google ScholarPubMed
Hine, CM et al. (2008) Use of the Rad51 promoter for targeted anti-cancer therapy. Proceedings of the National Academy of Sciences of the United States of America 105, 2081020815.Google ScholarPubMed
Balacescu, O et al. (2014) Gene expression profiling reveals activation of the FA/BRCA pathway in advanced squamous cervical cancer with intrinsic resistance and therapy failure. BMC Cancer 14, 246.Google ScholarPubMed
Gachechiladze, M et al. (2017) RAD51 as a potential surrogate marker for DNA repair capacity in solid malignancies. International Journal of Cancer 141, 12861294.Google ScholarPubMed
Morrison, C et al. (2021) Expression levels of RAD51 inversely correlate with survival of glioblastoma patients. Cancers 13, 5358.Google ScholarPubMed
Chang, HHY et al. (2017) Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nature Reviews Molecular Cell Biology 18, 495506.Google ScholarPubMed
Chen, M et al. (2022) DNA Damage response evaluation provides novel insights for personalized immunotherapy in glioma. Frontiers in Immunology 13, 875648.Google ScholarPubMed
England, B et al. (2013) Current understanding of the role and targeting of tumor suppressor p53 in glioblastoma multiforme. Tumour Biology Journal 34, 20632074.Google ScholarPubMed
Sun, Q et al. (2019) Therapeutic implications of p53 status on cancer cell fate following exposure to ionizing radiation and the DNA-PK inhibitor M3814. Molecular Cancer Research 17, 24572468.Google ScholarPubMed
Young, LC et al. (2013) Kdm4b histone demethylase is a DNA damage response protein and confers a survival advantage following γ-irradiation. Journal of Biological Chemistry 288, 2137621388.Google ScholarPubMed
Sule, A et al. (2021) Targeting IDH1/2 mutant cancers with combinations of ATR and PARP inhibitors. NAR Cancer 3, zcab018.Google ScholarPubMed
Eich, M et al. (2013) Contribution of ATM and ATR to the resistance of glioblastoma and malignant melanoma cells to the methylating anticancer drug temozolomide. Molecular Cancer Therapy 12, 25292540.Google Scholar
Behrooz, AB et al. (2022) Wnt and PI3K/Akt/mTOR survival pathways as therapeutic targets in glioblastoma. International Journal of Molecular Sciences 23, 353.Google Scholar
Benitez, JA et al. (2017) PTEN regulates glioblastoma oncogenesis through chromatin-associated complexes of DAXX and histone H3.3. Nature Communication 8, 15223.Google ScholarPubMed
McCabe, N et al. (2015) Mechanistic rationale to target PTEN-deficient tumor cells with inhibitors of the DNA damage response kinase ATM. Cancer Research 75, 21592165.Google ScholarPubMed
Turchick, A et al. (2019) Synthetic lethality of a cell-penetrating anti-RAD51 antibody in PTEN-deficient melanoma and glioma cells. Oncotarget 10, 12721283.Google ScholarPubMed
He, J et al. (2015) PTEN regulates DNA replication progression and stalled fork recovery. Nature Communication 6, 7620.Google ScholarPubMed
Rein, HL et al. (2021) RAD51 paralog function in replicative DNA damage and tolerance. Current Opinion in Genetics & Development 71, 8691.Google ScholarPubMed
Welsh, JW et al. (2009) Rad51 protein expression and survival in patients with glioblastoma multiforme. International Journal of Radiation Oncology - Biology - Physics 74, 12511255.Google ScholarPubMed
Pirlog, R et al. (2019) Proteomic advances in glial tumors through mass spectrometry approaches. Medicina 55, 412.Google ScholarPubMed
Susman, S et al. (2019) The role of p-Stat3 Y705 immunohistochemistry in glioblastoma prognosis. Diagnostic Pathology 14, 124.Google ScholarPubMed
Palla, VV et al. (2017) gamma-H2AX: can it be established as a classical cancer prognostic factor? Tumor Biology 39, 1010428317695931.Google ScholarPubMed
Nagelkerke, A and Span, PN (2016) Staining against phospho-H2AX (γ-H2AX) as a marker for DNA damage and genomic instability in cancer tissues and cells. Advances in Experimental Medicine and Biology 899, 110.Google ScholarPubMed
Jones, GN et al. (2018) pRAD50: a novel and clinically applicable pharmacodynamic biomarker of both ATM and ATR inhibition identified using mass spectrometry and immunohistochemistry. British Journal of Cancer 119, 12331243.Google ScholarPubMed
Martin, JG et al. (2021) Chemoproteomic profiling of covalent XPO1 inhibitors to assess target engagement and selectivity. Chembiochem 22, 21162123.Google ScholarPubMed
Birner, P et al. (2002) Prognostic relevance of p53 protein expression in glioblastoma. Oncology Reports 9, 703707.Google ScholarPubMed
Seol, HJ et al. (2011) Prognostic implications of the DNA damage response pathway in glioblastoma. Oncology Reports 26, 423430.Google ScholarPubMed
Majd, NK et al. (2021) The promise of DNA damage response inhibitors for the treatment of glioblastoma. Neuro-Oncol Advances 3, vdab015.Google ScholarPubMed
Ghelli Luserna di Rorà, A et al. (2020) A WEE1 family business: regulation of mitosis, cancer progression, and therapeutic target. Journal of Hematology Oncology 13, 126.Google ScholarPubMed
Śledzińska, P et al. (2021) Prognostic and predictive biomarkers in gliomas. International Journal of Molecular Sciences 22, 10373.Google ScholarPubMed
DeOcesano-Pereira, C et al. (2020) Emerging roles and potential applications of non-coding RNAs in glioblastoma. International Journal of Molecular Sciences 21, E2611.Google ScholarPubMed
Banelli, B et al. (2017) MicroRNA in glioblastoma: an overview. International Journal of Genomics 2017, 7639084.Google ScholarPubMed
Wu, P et al. (2019) Lnc-TALC promotes O6-methylguanine-DNA methyltransferase expression via regulating the c-Met pathway by competitively binding with miR-20b-3p. Nature Communications 10, 2045.Google Scholar
Beylerli, O et al. (2022) Long noncoding RNAs as promising biomarkers in cancer. Non-Coding RNA Research 7, 6670.Google ScholarPubMed
Van Roosbroeck, K and Calin, GA (2017) Cancer hallmarks and microRNAs: the therapeutic connection. Advances in Cancer Research 135, 119149.Google ScholarPubMed
Paulmurugan, R et al. (2019) The protean world of non-coding RNAs in glioblastoma. The Journal of Molecular Medicine 97, 909925.Google ScholarPubMed
Lu, E et al. (2022) The mechanisms of current platinum anticancer drug resistance in the glioma. Current Pharmaceutical Design 28, 18631869.Google ScholarPubMed
Visser, H and Thomas, AD (2021) MicroRNAs and the DNA damage response: how is cell fate determined? DNA Repair 108, 103245.Google Scholar
Chen, M et al. (2021) Role of microRNAs in glioblastoma. Oncotarget 12, 17071723.Google ScholarPubMed
Costa, PM et al. (2015) MicroRNAs in glioblastoma: role in pathogenesis and opportunities for targeted therapies. CNS & Neurological Disorders - Drug Targets 14, 222238.Google ScholarPubMed
Yadav, B et al. (2021) LncRNAs associated with glioblastoma: from transcriptional noise to novel regulators with a promising role in therapeutics. Molecular Therapy Nucleic Acids 24, 728742.CrossRefGoogle ScholarPubMed
Winkle, M et al. (2021) Noncoding RNA therapeutics - challenges and potential solutions. Nature Reviews Drug Discovery 20, 629651.Google ScholarPubMed
Lei, Q et al. (2023) MicroRNA-based therapy for glioblastoma: opportunities and challenges. European Journal of Pharmacology 938, 175388.Google ScholarPubMed
Reda El Sayed, S et al. (2021) MicroRNA therapeutics in cancer: current advances and challenges. Cancers 13, 2680.Google ScholarPubMed
Shetty, K et al. (2022) Multifunctional nanocarriers for delivering siRNA and miRNA in glioblastoma therapy: advances in nanobiotechnology-based cancer therapy. 3 Biotech 12, 301.Google ScholarPubMed
Costa, PM et al. (2015) MiRNA-21 silencing mediated by tumor-targeted nanoparticles combined with sunitinib: a new multimodal gene therapy approach for glioblastoma. Journal of Controlled Release 207, 3139.Google ScholarPubMed
Lee, TJ et al. (2017) RNA nanoparticle-based targeted therapy for glioblastoma through inhibition of oncogenic miR-21. Molecular Therapy 25, 15441555.Google ScholarPubMed
Li, H et al. (2018) miR-519a enhances chemosensitivity and promotes autophagy in glioblastoma by targeting STAT3/Bcl2 signaling pathway. Journal of Hematology Oncology 11, 70.Google ScholarPubMed
Nan, Y et al. (2021) miRNA-451 regulates the NF-κB signaling pathway by targeting IKKβ to inhibit glioma cell growth. Cell Cycle 20, 1967–1177.Google ScholarPubMed
Mah, LJ et al. (2010) γH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 24, 679686.Google ScholarPubMed
Vinchure, OS et al. (2021) miR-490 suppresses telomere maintenance program and associated hallmarks in glioblastoma. Cellular and Molecular Life Sciences 78, 22992314.Google ScholarPubMed
Williams, AB and Schumacher, B (2016) P53 in the DNA-damage-repair process. Cold Spring Harbor Perspectives in Medicine 6, a026070.Google ScholarPubMed
Besse, A et al. (2016) MiR-338-5p sensitizes glioblastoma cells to radiation through regulation of genes involved in DNA damage response. Tumour Biology 37, 77197727.Google ScholarPubMed
Wang, W et al. (2017) DNA damage-induced nuclear factor-kappa B activation and its roles in cancer progression. Journal of Cancer Metastasis and Treatment 3, 4559.Google ScholarPubMed
Soubannier, V and Stifani, S (2017) NF-κB Signalling in glioblastoma. Biomedicines 5, E29.Google ScholarPubMed
Xu, RX et al. (2015) DNA damage-induced NF-κB activation in human glioblastoma cells promotes miR-181b expression and cell proliferation. Cellular Physiology and Biochemistry 35, 913925.Google ScholarPubMed
Li, W et al. (2014) miR-221/222 confers radioresistance in glioblastoma cells through activating Akt independent of PTEN status. Current Molecular Medicine 14, 185195.Google ScholarPubMed
Quintavalle, C et al. (2013) MiR-221/222 target the DNA methyltransferase MGMT in glioma cells. PLoS ONE 8, e74466.Google ScholarPubMed
O'Brien, J et al. (2018) Overview of microRNA biogenesis, mechanisms of actions, and circulation. Frontiers in Endocrinology 9, 402.Google ScholarPubMed
Sati, ISEE and Parhar, I (2021) MicroRNAs regulate cell cycle and cell death pathways in glioblastoma. International Journal of Molecular Sciences 22, 13550.Google ScholarPubMed
Peng, Y and Croce, CM (2016) The role of microRNAs in human cancer. Signal Transduction and Targeted Therapy 1, 15004.Google ScholarPubMed
Yin, J et al. (2021) Extracellular vesicles derived from hypoxic glioma stem-like cells confer temozolomide resistance on glioblastoma by delivering miR-30b-3p. Theranostics 11, 17631779.Google ScholarPubMed
Wong, STS et al. (2012) MicroRNA-21 inhibition enhances in vitro chemosensitivity of temozolomide-resistant glioblastoma cells. Anticancer Research 32, 28352841.Google ScholarPubMed
Wang, G et al. (2015) Targeting strategies on miRNA-21 and PDCD4 for glioblastoma. Archives of Biochemistry and Biophysics 580, 6474.Google ScholarPubMed
Aloizou, AM et al. (2020) The role of MiRNA-21 in gliomas: hope for a novel therapeutic intervention? Toxicology Reports 7, 15141530.Google ScholarPubMed
Chen, YY et al. (2018) Upregulation of miR-125b, miR-181d, and miR-221 predicts poor prognosis in MGMT promoter-unmethylated glioblastoma patients. American Journal of Clinical Pathology 149, 412417.Google ScholarPubMed
Wang, P et al. (2020) The HIF1α/HIF2α-miR210-3p network regulates glioblastoma cell proliferation, dedifferentiation and chemoresistance through EGF under hypoxic conditions. Cell Death Disease 11, 992.Google ScholarPubMed
Munoz, JL et al. (2015) Temozolomide resistance in glioblastoma occurs by miRNA-9-targeted PTCH1, independent of sonic hedgehog level. Oncotarget 6, 11901201.Google ScholarPubMed
Zhang, J et al. (2020) Inhibition of miR-1193 leads to synthetic lethality in glioblastoma multiforme cells deficient of DNA-PKcs. Cell Death Disease 11, 602.Google ScholarPubMed
Wang, L et al. (2014) MiR-143 acts as a tumor suppressor by targeting N-RAS and enhances temozolomide-induced apoptosis in glioma. Oncotarget 5, 54165427.Google ScholarPubMed
Berthois, Y et al. (2014) Differential expression of miR200a-3p and miR21 in grade II-III and grade IV gliomas: evidence that miR200a-3p is regulated by O6-methylguanine methyltransferase and promotes temozolomide responsiveness. Cancer Biology Therapy 15, 938950.Google ScholarPubMed
Xiao, S et al. (2016) miR-29c contribute to glioma cells temozolomide sensitivity by targeting O6-methylguanine-DNA methyltransferases indirectely. Oncotarget 7, 5022950238.Google ScholarPubMed
Wu, H et al. (2014) MiR-136 modulates glioma cell sensitivity to temozolomide by targeting astrocyte elevated gene-1. Diagnostic Pathology 9, 173.Google ScholarPubMed
Liu, Q et al. (2015) miR-155 regulates glioma cells invasion and chemosensitivity by p38 isforms in vitro. Journal of Cellular Biochemistry 116, 12131221.Google ScholarPubMed
Zhen, L et al. (2016) MiR-10b decreases sensitivity of glioblastoma cells to radiation by targeting AKT. Journal of Biological Research 23, 14.Google ScholarPubMed
Guo, P et al. (2018) Upregulation of miR-96 promotes radioresistance in glioblastoma cells via targeting PDCD4. International Journal of Oncology 53, 15911600.Google ScholarPubMed
Guo, P et al. (2014) MiR-26a enhances the radiosensitivity of glioblastoma multiforme cells through targeting of ataxia-telangiectasia mutated. Experimental Cell Research 320, 200208.Google ScholarPubMed
Yan, D et al. (2010) Targeting DNA-PKcs and ATM with miR-101 sensitizes tumors to radiation. PLoS ONE 5, e11397.Google ScholarPubMed
He, X and Fan, S (2018) hsa-miR-212 modulates the radiosensitivity of glioma cells by targeting BRCA1. Oncology Reports 39, 977984.Google ScholarPubMed
DeSouza, PA et al. (2021) Long, noncoding RNA dysregulation in glioblastoma. Cancers 13, 1604.Google ScholarPubMed
Li, Z et al. (2019) Modulating lncRNA SNHG15/CDK6/miR-627 circuit by palbociclib, overcomes temozolomide resistance and reduces M2-polarization of glioma associated microglia in glioblastoma multiforme. Journal of Experimental & Clinical Cancer Research 38, 380.Google ScholarPubMed
Xu, C et al. (2022) lncRNA PRADX is a mesenchymal glioblastoma biomarker for cellular metabolism targeted therapy. Frontiers in Oncology 12, 888922.Google ScholarPubMed
Li, J et al. (2018) Targeting long noncoding RNA HMMR-AS1 suppresses and radiosensitizes glioblastoma. Neoplasia 20, 456466.Google ScholarPubMed
Yuan, E et al. (2022) Modulating glioblastoma chemotherapy response: evaluating long non-coding RNA effects on DNA damage response, glioma stem cell function, and hypoxic processes. Neuro-Oncology Advances 4, vdac119.Google ScholarPubMed
Zhang, Z et al. (2019) Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. Journal of Experimental & Clinical Cancer Research 38, 166.Google ScholarPubMed
Voce, DJ et al. (2019) Temozolomide treatment induces lncRNA MALAT1 in an NF-κB and p53 codependent manner in glioblastoma. Cancer Research 79, 25362548.Google Scholar
Shangguan, W et al. (2019) FoxD2-AS1 is a prognostic factor in glioma and promotes temozolomide resistance in a O6-methylguanine-DNA methyltransferase-dependent manner. Korean Journal of Physiology and Pharmacology 23, 475482.Google Scholar
Nie, E et al. (2021) TGF-β1 modulates temozolomide resistance in glioblastoma via altered microRNA processing and elevated MGMT. Neuro-Oncology 23, 435446.Google ScholarPubMed
Gong, R et al. (2021) Long noncoding RNA PVT1 promotes stemness and temozolomide resistance through miR-365/ELF4/SOX2 axis in glioma. Experimental Neurobiology 30, 244255.Google ScholarPubMed
Yan, Y et al. (2019) Novel function of lncRNA ADAMTS9-AS2 in promoting temozolomide resistance in glioblastoma via upregulating the FUS/MDM2 ubiquitination axis. Frontiers in Cell and Developmental Biology 7, 217.Google ScholarPubMed
Liao, Y et al. (2017) LncRNA CASC2 interacts with miR-181a to modulate glioma growth and resistance to TMZ through PTEN pathway. Journal of Cellular Biochemistry 118, 18891899.Google ScholarPubMed
Ding, J et al. (2020) lncRNA CCAT2 enhanced resistance of glioma cells against chemodrugs by disturbing the normal function of miR-424. OncoTargets Therapy 13, 14311445.CrossRefGoogle ScholarPubMed
Yuan, Z et al. (2020) Exosome-mediated transfer of long noncoding RNA HOTAIR regulates temozolomide resistance by miR-519a-3p/RRM1 axis in glioblastoma. Cancer Biotherapy and Radiopharmaceuticals. doi: 10.1089/cbr.2019.3499.Google ScholarPubMed
Chen, M et al. (2020) NCK1-AS1 Increases drug resistance of glioma cells to temozolomide by modulating miR-137/TRIM24. Cancer Biotherapy and Radiopharmaceuticals 35, 101108.Google ScholarPubMed
Liu, B et al. (2020) LncRNA SOX2OT promotes temozolomide resistance by elevating SOX2 expression via ALKBH5-mediated epigenetic regulation in glioblastoma. Cell Death Disease 11, 384.Google ScholarPubMed
Du, P et al. (2017) LncRNA-XIST interacts with miR-29c to modulate the chemoresistance of glioma cell to TMZ through DNA mismatch repair pathway. Bioscience Reports 37, BSR20170696.Google ScholarPubMed
Dai, X et al. (2019) AHIF promotes glioblastoma progression and radioresistance via exosomes. International Journal of Oncology 54, 261270.Google ScholarPubMed
Tang, G et al. (2021) lncRNA LINC01057 promotes mesenchymal differentiation by activating NF-κB signaling in glioblastoma. Cancer Letters 498, 152164.Google ScholarPubMed
Zheng, J et al. (2020) Linc-RA1 inhibits autophagy and promotes radioresistance by preventing H2Bub1/USP44 combination in glioma cells. Cell Death Disease 11, 758.Google ScholarPubMed
Hao, Z et al. (2019) Circular RNAs: functions and prospects in glioma. Journal of Molecular Neurosciences 67, 7281.Google ScholarPubMed
Rybak-Wolf, A et al. (2015) Circular RNAs in the mammalian brain are highly abundant, conserved, and dynamically expressed. Molecular Cell 58, 870885.Google ScholarPubMed
Sun, J et al. (2020) Functions and clinical significance of circular RNAs in glioma. Molecular Cancer 19, 34.Google ScholarPubMed
Wang, X et al. (2021) Identification of low-dose radiation-induced exosomal circ-METRN and miR-4709-3p/GRB14/PDGFRα pathway as a key regulatory mechanism in glioblastoma progression and radioresistance: functional validation and clinical theranostic significance. International Journal of Biological Sciences 17, 10611078.Google ScholarPubMed
Lou, J et al. (2020) Circular RNA CDR1as disrupts the p53/MDM2 complex to inhibit Gliomagenesis. Molecular Cancer 19, 138.Google ScholarPubMed
Wang, HX et al. (2018) Expression profile of circular RNAs in IDH-wild type glioblastoma tissues. Clinical Neurology and Neurosurgery 171, 168173.Google ScholarPubMed
Song, J et al. (2022) A novel protein encoded by ZCRB1-induced circHEATR5B suppresses aerobic glycolysis of GBM through phosphorylation of JMJD5. Journal of Experimental & Clinical Cancer Research 41, 171.Google ScholarPubMed
Jiang, Y et al. (2022) CircLRFN5 inhibits the progression of glioblastoma via PRRX2/GCH1 mediated ferroptosis. Journal of Experimental & Clinical Cancer Research 41, 307.Google ScholarPubMed
Caron, MC et al. (2019) Poly(ADP-ribose) polymerase-1 antagonizes DNA resection at double-strand breaks. Nature Communications 10, 2954.Google ScholarPubMed
Pascal, JM (2018) The comings and goings of PARP-1 in response to DNA damage. DNA Repair 71, 177182.Google ScholarPubMed
Sim, HW et al. (2022) PARP Inhibitors in glioma: a review of therapeutic opportunities. Cancers 14, 1003.Google ScholarPubMed
Murnyák, B et al. (2017) PARP1 Expression and its correlation with survival is tumour molecular subtype dependent in glioblastoma. Oncotarget 8, 4634846362.Google ScholarPubMed
Gourley, C et al. (2019) Moving from poly (ADP-ribose) polymerase inhibition to targeting DNA repair and DNA damage response in cancer therapy. Journal of Clinical Oncology 37, 22572269.Google ScholarPubMed
Kazlauskas, A et al. (2019) Isocytosine deaminase Vcz as a novel tool for the prodrug cancer therapy. BMC Cancer 19, 197.Google ScholarPubMed
Hanna, C et al. (2020) Pharmacokinetics, safety, and tolerability of olaparib and temozolomide for recurrent glioblastoma: results of the phase I OPARATIC trial. Neuro-Oncology 22, 18401850.Google ScholarPubMed
Sim, HW et al. (2021) A randomized phase II trial of veliparib, radiotherapy, and temozolomide in patients with unmethylated MGMT glioblastoma: the VERTU study. Neuro-Oncology 23, 17361749.Google ScholarPubMed
Cimprich, KA and Cortez, D (2008) ATR: an essential regulator of genome integrity. Nature Review Molecular Cellular Biology 9, 616627.Google ScholarPubMed
Frosina, G et al. (2019) The efficacy and toxicity of ATM inhibition in glioblastoma initiating cells-driven tumor models. Critical Reviews in Oncology and Hematology 138, 214222.Google ScholarPubMed
Jucaite, A et al. (2020) Brain exposure of the ATM inhibitor AZD1390 in humans—a positron emission tomography study. Neuro-Oncology 23, 687696.Google Scholar
Vecchio, D et al. (2014) Predictability, efficacy and safety of radiosensitization of glioblastoma-initiating cells by the ATM inhibitor KU-60019. International Journal of Cancer 135, 479491.Google ScholarPubMed
Green, AL et al. (2015) Preclinical antitumor efficacy of selective exportin 1 inhibitors in glioblastoma. Neuro-Oncology 17, 697707.Google ScholarPubMed
Degorre, C et al. (2021) Bench to bedside radiosensitizer development strategy for newly diagnosed glioblastoma. Radiation Oncology 16, 191.Google ScholarPubMed
Lassman, AB et al. (2022) A phase II study of the efficacy and safety of oral selinexor in recurrent glioblastoma. Clinical Cancer Research 28, 452460.Google ScholarPubMed
Wu, S et al. (2018) Activation of WEE1 confers resistance to PI3 K inhibition in glioblastoma. Neuro-Oncology 20, 7891.Google Scholar
Sanai, N et al. (2018) Phase 0 trial of AZD1775 in first-recurrence glioblastoma patients. Clinical Cancer Research 24, 38203828.Google Scholar
Lewis, CW et al. (2019) Upregulation of Myt1 promotes acquired resistance of cancer cells to Wee1 inhibition. Cancer Research 79, 59715985.Google ScholarPubMed