Neuromodulation for Brain Tumors: Myth or Reality? A Narrative Review

In recent years, research on brain cancers has turned towards the study of the interplay between the tumor and its host, the normal brain. Starting from the establishment of a parallelism between neurogenesis and gliomagenesis, the influence of neuronal activity on the development of brain tumors, particularly gliomas, has been partially unveiled. Notably, direct electrochemical synapses between neurons and glioma cells have been identified, paving the way for new approaches for the cure of brain cancers. Since this novel field of study has been defined “cancer neuroscience”, anticancer therapeutic approaches exploiting these discoveries can be referred to as “cancer neuromodulation”. In the present review, we provide an up-to-date description of the novel findings and of the therapeutic neuromodulation perspectives in cancer neuroscience. We focus both on more traditional oncologic approaches, aimed at modulating the major pathways involved in cancer neuroscience through drugs or genetic engineering techniques, and on electric stimulation proposals; the latter is at the cutting-edge of neuro-oncology.

[1]  Xing-jian Lin,et al.  Deep brain stimulation improves central nervous system inflammation in Parkinson's disease: Evidence and perspectives , 2023, CNS neuroscience & therapeutics.

[2]  F. Wagner,et al.  Therapy Resistance of Glioblastoma in Relation to the Subventricular Zone: What Is the Role of Radiotherapy? , 2023, Cancers.

[3]  G. Pavesi,et al.  Efficacy and Tolerability of Perampanel in Brain Tumor-Related Epilepsy: A Systematic Review , 2023, Biomedicines.

[4]  J. Suckling,et al.  Transcriptomic and connectomic correlates of differential spatial patterning among gliomas , 2022, Brain : a journal of neurology.

[5]  F. Winkler,et al.  Insights and opportunities at the crossroads of cancer and neuroscience , 2022, Nature Cell Biology.

[6]  S. Casas-Tintó,et al.  Synaptic components are required for glioblastoma progression in Drosophila , 2022, PLoS genetics.

[7]  S. May,et al.  Genome-Wide Expression and Anti-Proliferative Effects of Electric Field Therapy on Pediatric and Adult Brain Tumors , 2022, International journal of molecular sciences.

[8]  Shu-Hong Huang,et al.  NLGN3 Upregulates Expression of ADAM10 to Promote the Cleavage of NLGN3 via Activating the LYN Pathway in Human Gliomas , 2021, Frontiers in Cell and Developmental Biology.

[9]  Á. Pascual-Leone,et al.  Personalised, image-guided, noninvasive brain stimulation in gliomas: Rationale, challenges and opportunities , 2021, EBioMedicine.

[10]  F. Liu,et al.  Neuronal-driven glioma growth requires Gαi1 and Gαi3 , 2021, Theranostics.

[11]  Zaixu Cui,et al.  Structural connectome quantifies tumour invasion and predicts survival in glioblastoma patients , 2021, bioRxiv.

[12]  G. Reifenberger,et al.  The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. , 2021, Neuro-oncology.

[13]  J. Huguenard,et al.  NF1 mutation drives neuronal activity-dependent initiation of optic glioma , 2021, Nature.

[14]  Dong Zhou,et al.  Validation of the functions and prognostic values of synapse-associated proteins in lower-grade glioma , 2020, Bioscience reports.

[15]  W. Glac,et al.  Subthalamic Deep Brain Stimulation Affects Plasma Corticosterone Concentration and Peripheral Immunity Changes in Rat Model of Parkinson’s Disease , 2020, Journal of Neuroimmune Pharmacology.

[16]  H. Friedman,et al.  Management of glioblastoma: State of the art and future directions. , 2020, CA: a cancer journal for clinicians.

[17]  L. Ricci-Vitiani,et al.  Rapid and Efficient Invasion Assay of Glioblastoma in Human Brain Organoids. , 2020, Cell reports.

[18]  A. Yool,et al.  Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion , 2020, Frontiers in Pharmacology.

[19]  Hesheng Liu,et al.  Resting-state fMRI Detects Alterations in Whole Brain Connectivity Related to Tumor Biology in Glioma Patients. , 2020, Neuro-oncology.

[20]  H. Monyer,et al.  Emerging intersections between neuroscience and glioma biology , 2019, Nature Neuroscience.

[21]  Kexian Zhang,et al.  Systemic screening identifies GABRD, a subunit gene of GABAA receptor as a prognostic marker in adult IDH wild-type diffuse low-grade glioma. , 2019, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[22]  Shawn M. Gillespie,et al.  Electrical and synaptic integration of glioma into neural circuits , 2019, Nature.

[23]  Pengfei Gao,et al.  NLGN3 promotes neuroblastoma cell proliferation and growth through activating PI3K/AKT pathway. , 2019, European journal of pharmacology.

[24]  A. Korshunov,et al.  Acyl-CoA-Binding Protein Drives Glioblastoma Tumorigenesis by Sustaining Fatty Acid Oxidation. , 2019, Cell metabolism.

[25]  Chen Wang,et al.  Evaluation of serum extracellular vesicles as noninvasive diagnostic markers of glioma , 2019, Theranostics.

[26]  A. Alessandrini,et al.  Glutamate Receptors and Glioblastoma Multiforme: An Old “Route” for New Perspectives , 2019, International journal of molecular sciences.

[27]  T. H. Nguyen,et al.  Global, regional, and national burden of brain and other CNS cancer, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016 , 2019, The Lancet Neurology.

[28]  Shi-ting Li,et al.  CA10 and CA11 negatively regulate neuronal activity‐dependent growth of gliomas , 2019, Molecular oncology.

[29]  R. Köhling,et al.  AMPA receptor antagonist perampanel affects glioblastoma cell growth and glutamate release in vitro , 2019, PloS one.

[30]  R. Nicoll,et al.  Isoform-specific cleavage of neuroligin-3 reduces synapse strength , 2018, Molecular Psychiatry.

[31]  A. Hillebrand,et al.  Oscillatory brain activity associates with neuroligin-3 expression and predicts progression free survival in patients with diffuse glioma , 2018, Journal of Neuro-Oncology.

[32]  Rui Liu,et al.  Glioblastoma recurrence correlates with NLGN3 levels , 2018, Cancer medicine.

[33]  S. Schmid,et al.  Preclinical outcomes of Intratumoral Modulation Therapy for glioblastoma , 2018, Scientific Reports.

[34]  F. Lieberman,et al.  Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial , 2017, JAMA.

[35]  A. Olivi,et al.  The clinical value of patient-derived glioblastoma tumorspheres in predicting treatment response , 2017, Neuro-oncology.

[36]  T. Harkany,et al.  Endogenous GABAA receptor activity suppresses glioma growth , 2017, Oncogene.

[37]  J. Custódio,et al.  Ionotropic glutamate receptor antagonists and cancer therapy: time to think out of the box? , 2017, Cancer Chemotherapy and Pharmacology.

[38]  Parag Mallick,et al.  Neuronal Activity Promotes Glioma Growth through Neuroligin-3 Secretion , 2015, Cell.

[39]  L. Ricci-Vitiani,et al.  Type-3 metabotropic glutamate receptors regulate chemoresistance in glioma stem cells, and their levels are inversely related to survival in patients with malignant gliomas , 2012, Cell Death and Differentiation.

[40]  P. Gutin,et al.  NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. , 2012, European journal of cancer.

[41]  H. Monyer,et al.  Diazepam binding inhibitor promotes progenitor proliferation in the postnatal SVZ by reducing GABA signaling. , 2012, Cell stem cell.

[42]  J. Laterra,et al.  Neuro-oncology: Unmasking the multiforme in glioblastoma , 2010, Nature Reviews Neurology.

[43]  T. Mikkelsen,et al.  Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[44]  M. Passafaro,et al.  The GluR2 subunit inhibits proliferation by inactivating Src‐MAPK signalling and induces apoptosis by means of caspase 3/6‐dependent activation in glioma cells , 2009, The European journal of neuroscience.

[45]  L. Ricci-Vitiani,et al.  Cancer Stem Cell Analysis and Clinical Outcome in Patients with Glioblastoma Multiforme , 2008, Clinical Cancer Research.

[46]  L. Ricci-Vitiani,et al.  Type-3 metabotropic glutamate receptors negatively modulate bone morphogenetic protein receptor signaling and support the tumourigenic potential of glioma-initiating cells , 2008, Neuropharmacology.

[47]  Mitchel S Berger,et al.  Neural stem cells and the origin of gliomas. , 2005, The New England journal of medicine.

[48]  A. Bordey,et al.  Nonsynaptic GABA signaling in postnatal subventricular zone controls proliferation of GFAP-expressing progenitors , 2005, Nature Neuroscience.

[49]  R. Henkelman,et al.  Identification of human brain tumour initiating cells , 2004, Nature.

[50]  Ugo Orfanelli,et al.  Isolation and Characterization of Tumorigenic, Stem-like Neural Precursors from Human Glioblastoma , 2004, Cancer Research.

[51]  Y. Yoshida,et al.  Blockage of Ca2+-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells , 2002, Nature Medicine.

[52]  E. Holland,et al.  Glioblastoma multiforme: the terminator. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[53]  S. Weiss,et al.  Generation of neurons and astrocytes from isolated cells of the adult mammalian central nervous system. , 1992, Science.

[54]  S. Schmid,et al.  In Vitro Validation of Intratumoral Modulation Therapy for Glioblastoma. , 2016, Anticancer research.

[55]  A. Álvarez-Buylla,et al.  The heterogeneity of adult neural stem cells and the emerging complexity of their niche. , 2008, Cold Spring Harbor symposia on quantitative biology.