Drug resistance in glioblastoma: are persisters the key to therapy?

Glioblastoma (GBM) represents the main form of brain tumors in adults, and one of the most aggressive cancers overall. The treatment of GBM is a combination of surgery (when possible), chemotherapy (usually Temozolomide, TMZ) and radiotherapy (RT). However, despite this heavy treatment, GBM invariably recur and the median length of survival following diagnosis is 12 to 15 months, with less than 10% of people surviving longer than five years. GBM is extremely resistant to most treatments because of its heterogeneous nature, which is associated with extreme clonal plasticity and the presence of cancer stem cells, refractory to TMZ- and RT-induced cell death. In this review, we explore the mechanisms by which cancer cells, and especially GBM, can acquire resistance to treatment. We describe and discuss the concept of persister/tolerant cells that precede and/or accompany the acquisition of resistance. Persister/tolerant cells are cancer cells that are not eliminated by treatment(s) because of different mechanisms ranging from dormancy/quiescence to senescence. We discuss the possibility of targeting these mechanisms in new therapeutic regimen.

[1]  H. Schaider,et al.  Epigenetics and metabolism at the crossroads of stress-induced plasticity, stemness and therapeutic resistance in cancer , 2020, Theranostics.

[2]  Raymond B. Runyan,et al.  Guidelines and definitions for research on epithelial–mesenchymal transition , 2020, Nature Reviews Molecular Cell Biology.

[3]  G. Dunn,et al.  A review of glioblastoma immunotherapy , 2020, Journal of Neuro-Oncology.

[4]  K. Polyak,et al.  Intratumor Heterogeneity: The Rosetta Stone of Therapy Resistance. , 2020, Cancer cell.

[5]  J. Barciszewski,et al.  Revealing the epigenetic effect of temozolomide on glioblastoma cell lines in therapeutic conditions , 2020, PloS one.

[6]  Y. You,et al.  DNA-methylation-mediated activating of lncRNA SNHG12 promotes temozolomide resistance in glioblastoma , 2020, Molecular Cancer.

[7]  P. Marquet,et al.  Impairing temozolomide resistance driven by glioma stem‐like cells with adjuvant immunotherapy targeting O‐acetyl GD2 ganglioside , 2020, International journal of cancer.

[8]  G. F. Calvo,et al.  Identification of a transient state during the acquisition of temozolomide resistance in glioblastoma , 2020, Cell Death & Disease.

[9]  P. Cartron,et al.  Diuron exposure and Akt overexpression promote glioma formation through DNA hypomethylation , 2019, Clinical Epigenetics.

[10]  J. D. de Groot,et al.  Immune biology of glioma associated macrophages and microglia: Functional and therapeutic implications. , 2019, Neuro-oncology.

[11]  Neil Vasan,et al.  A view on drug resistance in cancer , 2019, Nature.

[12]  M. Neunlist,et al.  Tumor cells hijack enteric glia to activate colon cancer stem cells and stimulate tumorigenesis , 2019, EBioMedicine.

[13]  G. Lucignani,et al.  Intracellular Redox-Balance Involvement in Temozolomide Resistance-Related Molecular Mechanisms in Glioblastoma , 2019, Cells.

[14]  X. Saulquin,et al.  NKG2D Controls Natural Reactivity of Vγ9Vδ2 T Lymphocytes against Mesenchymal Glioblastoma Cells , 2019, Clinical Cancer Research.

[15]  Y. Assaraf,et al.  The multi-factorial nature of clinical multidrug resistance in cancer. , 2019, Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy.

[16]  T. Tenson,et al.  Slow growth causes bacterial persistence , 2019, Science Signaling.

[17]  Nader Sanai,et al.  A Phase 0 Trial of Ribociclib in Recurrent Glioblastoma Patients Incorporating a Tumor Pharmacodynamic- and Pharmacokinetic-Guided Expansion Cohort , 2019, Clinical Cancer Research.

[18]  H. Azari,et al.  Modulation of temozolomide dose differentially affects T-cell response to immune checkpoint inhibition. , 2019, Neuro-oncology.

[19]  J. Rich,et al.  Glioblastoma stem cells: lessons from the tumor hierarchy in a lethal cancer , 2019, Genes & development.

[20]  L. Cerchia,et al.  Proneural-Mesenchymal Transition: Phenotypic Plasticity to Acquire Multitherapy Resistance in Glioblastoma , 2019, International journal of molecular sciences.

[21]  A. Hueber,et al.  TRAIL and FasL Functions in Cancer and Autoimmune Diseases: Towards an Increasing Complexity , 2019, Cancers.

[22]  Jiandong Chen,et al.  Interaction between p53 N terminus and core domain regulates specific and nonspecific DNA binding , 2019, Proceedings of the National Academy of Sciences.

[23]  J. Collins,et al.  Definitions and guidelines for research on antibiotic persistence , 2019, Nature Reviews Microbiology.

[24]  P. Cartron,et al.  Dormant, quiescent, tolerant and persister cells: Four synonyms for the same target in cancer , 2019, Biochemical pharmacology.

[25]  C. Blanpain,et al.  EMT Transition States during Tumor Progression and Metastasis. , 2019, Trends in cell biology.

[26]  Piyush B. Gupta,et al.  Phenotypic Plasticity: Driver of Cancer Initiation, Progression, and Therapy Resistance. , 2019, Cell stem cell.

[27]  Daniela Matei,et al.  Targeting Cancer Stemness in the Clinic: From Hype to Hope. , 2019, Cell stem cell.

[28]  M. Christmann,et al.  Targeting anticancer drug-induced senescence in glioblastoma therapy , 2018, Oncotarget.

[29]  J. Sotelo,et al.  Autophagic and Apoptotic Pathways as Targets for Chemotherapy in Glioblastoma , 2018, International journal of molecular sciences.

[30]  D. Mitchell,et al.  Temozolomide for immunomodulation in the treatment of glioblastoma , 2018, Neuro-oncology.

[31]  M. Christmann,et al.  Temozolomide Induces Senescence and Repression of DNA Repair Pathways in Glioblastoma Cells via Activation of ATR-CHK1, p21, and NF-κB. , 2018, Cancer research.

[32]  R. Weinberg,et al.  New insights into the mechanisms of epithelial–mesenchymal transition and implications for cancer , 2018, Nature Reviews Molecular Cell Biology.

[33]  T. Garrett,et al.  Infiltrative and drug‐resistant slow‐cycling cells support metabolic heterogeneity in glioblastoma , 2018, The EMBO journal.

[34]  A. Heimberger,et al.  Cell surface vimentin-targeted monoclonal antibody 86C increases sensitivity to temozolomide in glioma stem cells. , 2018, Cancer letters.

[35]  P. Cartron,et al.  miR-370-3p Is a Therapeutic Tool in Anti-glioblastoma Therapy but Is Not an Intratumoral or Cell-free Circulating Biomarker , 2018, Molecular therapy. Nucleic acids.

[36]  Matthew A. Taylor,et al.  Targeting autophagy for combating chemoresistance and radioresistance in glioblastoma , 2018, Apoptosis.

[37]  I. Ulasov,et al.  Autophagy in glioma cells: An identity crisis with a clinical perspective. , 2018, Cancer letters.

[38]  Ji-Yong Um,et al.  Human glioblastoma arises from subventricular zone cells with low-level driver mutations , 2018, Nature.

[39]  L. Ricci-Vitiani,et al.  Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis , 2018, Cell Death & Disease.

[40]  J. Sarkaria,et al.  A novel enhancer regulates MGMT expression and promotes temozolomide resistance in glioblastoma , 2018, Nature Communications.

[41]  X. Breakefield,et al.  Multidimensional communication in the microenvirons of glioblastoma , 2018, Nature Reviews Neurology.

[42]  T. Mikkelsen,et al.  A Distinct DNA Methylation Shift in a Subset of Glioma CpG Island Methylator Phenotypes during Tumor Recurrence , 2018, Cell reports.

[43]  R. Molenaar,et al.  The hypoxic peri-arteriolar glioma stem cell niche, an integrated concept of five types of niches in human glioblastoma. , 2018, Biochimica et biophysica acta. Reviews on cancer.

[44]  H. Kearney,et al.  Temporal stability of MGMT promoter methylation in glioblastoma patients undergoing STUPP protocol , 2018, Journal of Neuro-Oncology.

[45]  J. Błasiak,et al.  An Interplay between Senescence, Apoptosis and Autophagy in Glioblastoma Multiforme—Role in Pathogenesis and Therapeutic Perspective , 2018, International journal of molecular sciences.

[46]  S. Fulda Cell death-based treatment of glioblastoma , 2018, Cell Death & Disease.

[47]  G. Ying,et al.  Glutathione reductase mediates drug resistance in glioblastoma cells by regulating redox homeostasis , 2018, Journal of neurochemistry.

[48]  H. Schaider,et al.  Tumor cell-intrinsic phenotypic plasticity facilitates adaptive cellular reprogramming driving acquired drug resistance , 2017, Journal of Cell Communication and Signaling.

[49]  P. Giresi,et al.  Repression of Stress-Induced LINE-1 Expression Protects Cancer Cell Subpopulations from Lethal Drug Exposure. , 2017, Cancer cell.

[50]  R. Teusan,et al.  Efficient Mitochondrial Glutamine Targeting Prevails Over Glioblastoma Metabolic Plasticity , 2017, Clinical Cancer Research.

[51]  M. Saparbaev,et al.  Aberrant base excision repair pathway of oxidatively damaged DNA: Implications for degenerative diseases☆ , 2017, Free radical biology & medicine.

[52]  M. Pistillo,et al.  Small molecules targeting histone demethylase genes (KDMs) inhibit growth of temozolomide-resistant glioblastoma cells , 2017, Oncotarget.

[53]  Shawn M. Gillespie,et al.  Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. , 2017, Cell stem cell.

[54]  D. Turnbull,et al.  Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells , 2016, Neuro-oncology.

[55]  E. Holland,et al.  Suppression of autophagy impedes glioblastoma development and induces senescence , 2016, Autophagy.

[56]  Sang Y Lee Temozolomide resistance in glioblastoma multiforme , 2016, Genes & diseases.

[57]  Hyunggee Kim,et al.  Irradiation induces glioblastoma cell senescence and senescence-associated secretory phenotype , 2016, Tumor Biology.

[58]  Bruce A. Posner,et al.  Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells , 2016, Nature Communications.

[59]  Ji-hua Zhong,et al.  RUNX1-Evi-1 fusion gene inhibited differentiation and apoptosis in myelopoiesis: an in vivo study , 2015, BMC Cancer.

[60]  N. Sharpless,et al.  Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice , 2015, Nature Medicine.

[61]  S. Zhong,et al.  Exosomes decrease sensitivity of breast cancer cells to adriamycin by delivering microRNAs , 2015, Tumor Biology.

[62]  Yuan Cheng,et al.  Targeting the Mitotic Catastrophe Signaling Pathway in Cancer , 2015, Mediators of inflammation.

[63]  D. Amadori,et al.  Exosome-mediated transfer of microRNAs within the tumor microenvironment and neuroblastoma resistance to chemotherapy. , 2015, Journal of the National Cancer Institute.

[64]  Z. Wang,et al.  MGMT promoter methylation in serum and cerebrospinal fluid as a tumor-specific biomarker of glioma. , 2015, Biomedical reports.

[65]  C. Pecqueur,et al.  D-2-Hydroxyglutarate does not mimic all the IDH mutation effects, in particular the reduced etoposide-triggered apoptosis mediated by an alteration in mitochondrial NADH , 2015, Cell Death and Disease.

[66]  F. Paris,et al.  Radiation-induced PGE2 sustains human glioma cell growth and survival through EGF signaling , 2015, Oncotarget.

[67]  M. Selak,et al.  Mitochondrial energy metabolism and apoptosis regulation in glioblastoma , 2015, Brain Research.

[68]  N. Salomonis,et al.  Reactive oxygen species-mediated therapeutic response and resistance in glioblastoma , 2015, Cell Death and Disease.

[69]  P. Lowenstein,et al.  Temozolomide Does Not Impair Gene Therapy-Mediated Antitumor Immunity in Syngeneic Brain Tumor Models , 2014, Clinical Cancer Research.

[70]  C. Pecqueur,et al.  Control of glioma cell death and differentiation by PKM2–Oct4 interaction , 2014, Cell Death and Disease.

[71]  L. Oliver,et al.  Bak and Mcl-1 are essential for Temozolomide induced cell death in human glioma , 2014, Oncotarget.

[72]  Philippe Juin,et al.  Decoding and unlocking the BCL-2 dependency of cancer cells , 2013, Nature Reviews Cancer.

[73]  L. O’Driscoll,et al.  Docetaxel-Resistance in Prostate Cancer: Evaluating Associated Phenotypic Changes and Potential for Resistance Transfer via Exosomes , 2012, PloS one.

[74]  S. Martin,et al.  Prognostic impact of the expression/phosphorylation of the BH3-only proteins of the BCL-2 family in glioblastoma multiforme , 2012, Cell Death and Disease.

[75]  S. Sengupta,et al.  Impact of Temozolomide on Immune Response during Malignant Glioma Chemotherapy , 2012, Clinical & developmental immunology.

[76]  M. Campone,et al.  Folate supplementation limits the tumourigenesis in rodent models of gliomagenesis. , 2012, European journal of cancer.

[77]  P. Naveilhan,et al.  Comparison of Spheroids Formed by Rat Glioma Stem Cells and Neural Stem Cells Reveals Differences in Glucose Metabolism and Promising Therapeutic Applications* , 2012, The Journal of Biological Chemistry.

[78]  C. Sommer,et al.  MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: a comparative study on astrocytoma and glioblastoma , 2010, International journal of cancer.

[79]  J. Mackey,et al.  Metabolic Modulation of Glioblastoma with Dichloroacetate , 2010, Science Translational Medicine.

[80]  Stephan Saikali,et al.  Prognostic value of O6-methylguanine-DNA methyltransferase status in glioblastoma patients, assessed by five different methods , 2010, Journal of Neuro-Oncology.

[81]  S. Gabriel,et al.  Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. , 2010, Cancer cell.

[82]  P. Cartron,et al.  Folate Supplementation Limits the Aggressiveness of Glioma via the Remethylation of DNA Repeats Element and Genes Governing Apoptosis and Proliferation , 2009, Clinical Cancer Research.

[83]  M. Berger,et al.  p53 Small-molecule inhibitor enhances temozolomide cytotoxic activity against intracranial glioblastoma xenografts. , 2008, Cancer research.

[84]  A. Guha,et al.  Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells , 2008, Nature Cell Biology.

[85]  Thomas Hundsberger,et al.  MGMT in primary and recurrent human glioblastomas after radiation and chemotherapy and comparison with p53 status and clinical outcome , 2008, International journal of cancer.

[86]  Mark W. Dewhirst,et al.  Glioma stem cells promote radioresistance by preferential activation of the DNA damage response , 2006, Nature.

[87]  P. Cartron,et al.  The small organic compound HA14-1 prevents Bcl-2 interaction with Bax to sensitize malignant glioma cells to induction of cell death. , 2006, Cancer research.

[88]  Martin J. van den Bent,et al.  Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. , 2005, The New England journal of medicine.

[89]  R. Mirimanoff,et al.  MGMT gene silencing and benefit from temozolomide in glioblastoma. , 2005, The New England journal of medicine.

[90]  M. Middleton,et al.  Temozolomide: a novel oral alkylating agent , 2001, Expert review of anticancer therapy.

[91]  P. Cartron,et al.  Expression Of Bcl-2, Bax and Bcl-xl in Human Gliomas: A Re-appraisal , 2001, Journal of Neuro-Oncology.

[92]  Scar,et al.  Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. , 2000, The New England journal of medicine.

[93]  A. Le,et al.  The Multifaceted Metabolism of Glioblastoma. , 2018, Advances in experimental medicine and biology.

[94]  G. Perkins,et al.  Finding a Panacea among combination cancer therapies. , 2012, Cancer research.

[95]  K. Lewis,et al.  Persister cells. , 2010, Annual review of microbiology.

[96]  W. Xiao The mitochondrial pathways of apoptosis , 2002 .