Access the full text.
Sign up today, get DeepDyve free for 14 days.
( Karanam N , Ding L , Aroumougame A , Story M . Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: implications for cancer therapy. Transl Res: J Lab Clin Med. 2020;217:33‐46.)
Karanam N , Ding L , Aroumougame A , Story M . Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: implications for cancer therapy. Transl Res: J Lab Clin Med. 2020;217:33‐46.Karanam N , Ding L , Aroumougame A , Story M . Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: implications for cancer therapy. Transl Res: J Lab Clin Med. 2020;217:33‐46., Karanam N , Ding L , Aroumougame A , Story M . Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: implications for cancer therapy. Transl Res: J Lab Clin Med. 2020;217:33‐46.
Yi Lin, Baoshi Chen (2021)
Case report: tumor-treating fields prolongs IDH-mutant anaplastic astrocytoma progression-free survival and pathological evolution to glioblastomaAnnals of Translational Medicine, 9
( Meletath S , Pavlick D , Brennan T , et al. Personalized treatment for a patient with a BRAF V600E mutation using dabrafenib and a tumor treatment fields device in a high‐grade glioma arising from ganglioglioma. J Nat Comprehens Cancer Network: JNCCN. 2016;14(11):1345‐1350.)
Meletath S , Pavlick D , Brennan T , et al. Personalized treatment for a patient with a BRAF V600E mutation using dabrafenib and a tumor treatment fields device in a high‐grade glioma arising from ganglioglioma. J Nat Comprehens Cancer Network: JNCCN. 2016;14(11):1345‐1350.Meletath S , Pavlick D , Brennan T , et al. Personalized treatment for a patient with a BRAF V600E mutation using dabrafenib and a tumor treatment fields device in a high‐grade glioma arising from ganglioglioma. J Nat Comprehens Cancer Network: JNCCN. 2016;14(11):1345‐1350., Meletath S , Pavlick D , Brennan T , et al. Personalized treatment for a patient with a BRAF V600E mutation using dabrafenib and a tumor treatment fields device in a high‐grade glioma arising from ganglioglioma. J Nat Comprehens Cancer Network: JNCCN. 2016;14(11):1345‐1350.
I. Vergote, R. Moos, L. Manso, E. Nieuwenhuysen, N. Concin, C. Sessa (2018)
Tumor Treating Fields in combination with paclitaxel in recurrent ovarian carcinoma: Results of the INNOVATE pilot study.Gynecologic oncology, 150 3
( Shi P , Tian J , Ulm B , et al. Tumor treating fields suppression of Ciliogenesis enhances temozolomide toxicity. Front Oncol. 2022;12:837589.35359402)
Shi P , Tian J , Ulm B , et al. Tumor treating fields suppression of Ciliogenesis enhances temozolomide toxicity. Front Oncol. 2022;12:837589.35359402Shi P , Tian J , Ulm B , et al. Tumor treating fields suppression of Ciliogenesis enhances temozolomide toxicity. Front Oncol. 2022;12:837589.35359402, Shi P , Tian J , Ulm B , et al. Tumor treating fields suppression of Ciliogenesis enhances temozolomide toxicity. Front Oncol. 2022;12:837589.35359402
(2017)
Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie
Guangrong Lu, Mayank Rao, P. Zhu, B. Liang, Rasheda El-Nazer, E. Fonkem, M. Bhattacharjee, Jay-Jiguang Zhu (2019)
Triple-drug Therapy With Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating Fields for Recurrent Glioblastoma: A Retrospective StudyFrontiers in Neurology, 10
( Lacouture M , Davis M , Elzinga G , et al. Characterization and management of dermatologic adverse events with the NovoTTF‐100A system, a novel anti‐mitotic electric field device for the treatment of recurrent glioblastoma. Semin Oncol. 2014;S1‐14:S1‐S14.)
Lacouture M , Davis M , Elzinga G , et al. Characterization and management of dermatologic adverse events with the NovoTTF‐100A system, a novel anti‐mitotic electric field device for the treatment of recurrent glioblastoma. Semin Oncol. 2014;S1‐14:S1‐S14.Lacouture M , Davis M , Elzinga G , et al. Characterization and management of dermatologic adverse events with the NovoTTF‐100A system, a novel anti‐mitotic electric field device for the treatment of recurrent glioblastoma. Semin Oncol. 2014;S1‐14:S1‐S14., Lacouture M , Davis M , Elzinga G , et al. Characterization and management of dermatologic adverse events with the NovoTTF‐100A system, a novel anti‐mitotic electric field device for the treatment of recurrent glioblastoma. Semin Oncol. 2014;S1‐14:S1‐S14.
N. Karanam, L. Ding, Asaithamby Aroumougame, M. Story (2019)
Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: Implications for cancer therapy.Translational research : the journal of laboratory and clinical medicine
( Chang E , Patel C , Pohling C , et al. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov. 2018;4:113.30534421)
Chang E , Patel C , Pohling C , et al. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov. 2018;4:113.30534421Chang E , Patel C , Pohling C , et al. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov. 2018;4:113.30534421, Chang E , Patel C , Pohling C , et al. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov. 2018;4:113.30534421
Eun Kim, Yunhui Jo, S. Sai, Munho Park, Jeong-Yub Kim, Jin Kim, Yeon-Joo Lee, Jae-Min Cho, S. Kwak, J. Baek, Youn Jeong, Jie-Young Song, M. Yoon, Sang-Gu Hwang (2019)
Tumor-treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cellsOncogene, 38
( Kim E , Jo Y , Sai S , et al. Tumor‐treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cells. Oncogene. 2019;38(39):6630‐6646.31375748)
Kim E , Jo Y , Sai S , et al. Tumor‐treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cells. Oncogene. 2019;38(39):6630‐6646.31375748Kim E , Jo Y , Sai S , et al. Tumor‐treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cells. Oncogene. 2019;38(39):6630‐6646.31375748, Kim E , Jo Y , Sai S , et al. Tumor‐treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cells. Oncogene. 2019;38(39):6630‐6646.31375748
Ronald Killiany, French Canadian, V. Belzil, P. Valdmanis, P. Dion, H. Daoud, E. Kabashi, A. Noreau, J. Gauthier, P. Hince, A. Desjarlais, J.-P Bouchard, L. Lacomblez, F. Salachas, P. Pradat, W. Camu, V. Meininger, N. Dupré, G. Rouleau, N. Ticozzi, V. Silani, A. Leclerc, P. Keagle, C. Gellera, A. Ratti, F. Taroni, T. Kwiatkowski, D. McKenna‐Yasek, P. Sapp, R. Brown, J. Landers, N. Scheinin, S. Aalto, J. Koikkalainen, J. Lötjönen, M. Karrasch, N. Kemppainen, M. Viitanen, K. Någren, S. Helin, M. Scheinin, J. Rinne, W. Jagust, S. Landau, L. Shaw, J. Trojanowski, R. Koeppe, E. Reiman, N. Foster, R. Petersen, M. Weiner, J. Price, F. Iwamoto, L. Abrey, K. Beal, P. Gutin, M. Rosenblum, V. Reuter, L. DeAngelis, A. Lassman, K. Jaeckle, K. Ballman, A. Furth, J. Buckner, H. Kurahashi, J.-W. Wang, A. Ishii, T. Kojima, S. Wakai, T. Kizawa, Y. Fujimoto, K. Kikkawa, K. Yoshimura, S. Hirose, R. Miller, C. Jackson, E. Kasarskis, J. England, D. Forshew, W. Johnston, S. Kalra, J. Katz, H. Mitsumoto, J. Rosenfeld, C. Shoesmith, M. Strong, S. Woolley, Stephen Sergay (2009)
Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma
M. Egeblad, Z. Werb (2002)
New functions for the matrix metalloproteinases in cancer progressionNature Reviews Cancer, 2
GBM: glioblastoma; rGBM: recurrence glioblastoma; ndGBM: newly-diagnosed glioblastoma; OS: overall survival; PFS: progression-free survival
( Mikic N , Poulsen F , Kristoffersen K , et al. Study protocol for OptimalTTF‐2: enhancing tumor treating fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi‐center, randomized, prospective, interventional trial. BMC Cancer. 2021;21(1):1010.34503460)
Mikic N , Poulsen F , Kristoffersen K , et al. Study protocol for OptimalTTF‐2: enhancing tumor treating fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi‐center, randomized, prospective, interventional trial. BMC Cancer. 2021;21(1):1010.34503460Mikic N , Poulsen F , Kristoffersen K , et al. Study protocol for OptimalTTF‐2: enhancing tumor treating fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi‐center, randomized, prospective, interventional trial. BMC Cancer. 2021;21(1):1010.34503460, Mikic N , Poulsen F , Kristoffersen K , et al. Study protocol for OptimalTTF‐2: enhancing tumor treating fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi‐center, randomized, prospective, interventional trial. BMC Cancer. 2021;21(1):1010.34503460
( Akbarnejad Z , Eskandary H , Dini L , et al. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low‐frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother. 2017;92:254‐264.28551545)
Akbarnejad Z , Eskandary H , Dini L , et al. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low‐frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother. 2017;92:254‐264.28551545Akbarnejad Z , Eskandary H , Dini L , et al. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low‐frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother. 2017;92:254‐264.28551545, Akbarnejad Z , Eskandary H , Dini L , et al. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low‐frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother. 2017;92:254‐264.28551545
M. Barreca, A. Stathis, P. Barraja, F. Bertoni (2020)
An overview on anti-tubulin agents for the treatment of lymphoma patients.Pharmacology & therapeutics
( Christofk H , Vander Heiden M , Harris M , et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452(7184):230‐233.18337823)
Christofk H , Vander Heiden M , Harris M , et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452(7184):230‐233.18337823Christofk H , Vander Heiden M , Harris M , et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452(7184):230‐233.18337823, Christofk H , Vander Heiden M , Harris M , et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452(7184):230‐233.18337823
S. Burri, V. Gondi, P. Brown, M. Mehta (2017)
The Evolving Role of Tumor Treating Fields in Managing GlioblastomaAmerican Journal of Clinical Oncology, 41
( Branter J , Estevez‐Cebrero M , Diksin M , et al. Genome‐wide expression and anti‐proliferative effects of electric field therapy on pediatric and adult brain tumors. Int J Mol Sci. 2022;23(4):1982.)
Branter J , Estevez‐Cebrero M , Diksin M , et al. Genome‐wide expression and anti‐proliferative effects of electric field therapy on pediatric and adult brain tumors. Int J Mol Sci. 2022;23(4):1982.Branter J , Estevez‐Cebrero M , Diksin M , et al. Genome‐wide expression and anti‐proliferative effects of electric field therapy on pediatric and adult brain tumors. Int J Mol Sci. 2022;23(4):1982., Branter J , Estevez‐Cebrero M , Diksin M , et al. Genome‐wide expression and anti‐proliferative effects of electric field therapy on pediatric and adult brain tumors. Int J Mol Sci. 2022;23(4):1982.
(2017)
ly low-frequency electromagnetic field (100Hz, 100G)
N. Karanam, Kalayarasan Srinivasan, L. Ding, Brock Sishc, D. Saha, M. Story (2017)
Tumor-treating fields elicit a conditional vulnerability to ionizing radiation via the downregulation of BRCA1 signaling and reduced DNA double-strand break repair capacity in non-small cell lung cancer cell linesCell Death & Disease, 8
Q. Ostrom, H. Gittleman, Paul Farah, A. Ondracek, Yanwen Chen, Yingli Wolinsky, N. Stroup, C. Kruchko, J. Barnholtz-Sloan (2013)
CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006-2010.Neuro-oncology, 15 Suppl 2
C. Patel, C. Beinat, Yuanyang Xie, E. Chang, S. Gambhir (2020)
Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [18F]DASA-23, a non-invasive probe of pyruvate kinase M2 (PKM2) expressionNeoplasia (New York, N.Y.), 23
( Rivoirard R , Vallard A , Langrand‐Escure J , et al. Thirty years of phase I radiochemotherapy trials: latest development. Eur J Cancer (Oxford, England: 1990). 2016;58:1‐7.)
Rivoirard R , Vallard A , Langrand‐Escure J , et al. Thirty years of phase I radiochemotherapy trials: latest development. Eur J Cancer (Oxford, England: 1990). 2016;58:1‐7.Rivoirard R , Vallard A , Langrand‐Escure J , et al. Thirty years of phase I radiochemotherapy trials: latest development. Eur J Cancer (Oxford, England: 1990). 2016;58:1‐7., Rivoirard R , Vallard A , Langrand‐Escure J , et al. Thirty years of phase I radiochemotherapy trials: latest development. Eur J Cancer (Oxford, England: 1990). 2016;58:1‐7.
( Burri S , Gondi V , Brown P , Mehta M . The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196.28832384)
Burri S , Gondi V , Brown P , Mehta M . The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196.28832384Burri S , Gondi V , Brown P , Mehta M . The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196.28832384, Burri S , Gondi V , Brown P , Mehta M . The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196.28832384
( Bernard‐Arnoux F , Lamure M , Ducray F , Aulagner G , Honnorat J , Armoiry X . The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136.27177573)
Bernard‐Arnoux F , Lamure M , Ducray F , Aulagner G , Honnorat J , Armoiry X . The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136.27177573Bernard‐Arnoux F , Lamure M , Ducray F , Aulagner G , Honnorat J , Armoiry X . The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136.27177573, Bernard‐Arnoux F , Lamure M , Ducray F , Aulagner G , Honnorat J , Armoiry X . The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136.27177573
( Jackson E , Barboriak D , Bidaut L , Meyer C . Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources. Transl Oncol. 2009;2(4):211‐215.19956380)
Jackson E , Barboriak D , Bidaut L , Meyer C . Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources. Transl Oncol. 2009;2(4):211‐215.19956380Jackson E , Barboriak D , Bidaut L , Meyer C . Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources. Transl Oncol. 2009;2(4):211‐215.19956380, Jackson E , Barboriak D , Bidaut L , Meyer C . Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources. Transl Oncol. 2009;2(4):211‐215.19956380
( Wick W . TTFields: where does all the skepticism come from? Neuro Oncol. 2016;18(3):303‐305.26917587)
Wick W . TTFields: where does all the skepticism come from? Neuro Oncol. 2016;18(3):303‐305.26917587Wick W . TTFields: where does all the skepticism come from? Neuro Oncol. 2016;18(3):303‐305.26917587, Wick W . TTFields: where does all the skepticism come from? Neuro Oncol. 2016;18(3):303‐305.26917587
( Kline‐Smith S , Walczak C . Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol Cell. 2004;15(3):317‐327.15304213)
Kline‐Smith S , Walczak C . Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol Cell. 2004;15(3):317‐327.15304213Kline‐Smith S , Walczak C . Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol Cell. 2004;15(3):317‐327.15304213, Kline‐Smith S , Walczak C . Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol Cell. 2004;15(3):317‐327.15304213
G. Elzinga, E. Wong (2014)
Resolution of Cystic Enhancement to Add-On Tumor Treating Electric Fields for Recurrent Glioblastoma after Incomplete Response to BevacizumabCase Reports in Neurology, 6
F. Bokstein, D. Blumenthal, D. Limon, C. Harosh, Z. Ram, R. Grossman (2020)
Concurrent Tumor Treating Fields (TTFields) and Radiation Therapy for Newly Diagnosed Glioblastoma: A Prospective Safety and Feasibility StudyFrontiers in Oncology, 10
Søs Holdgaard, V. Cianfanelli, Emanuela Pupo, M. Lambrughi, M. Lubas, Julie Nielsen, Susana Eibes, Emiliano Maiani, Lea Harder, Nicole Wesch, M. Foged, K. Maeda, F. Nazio, L. Ballina, V. Dötsch, A. Brech, Lisa Frankel, M. Jäättelä, F. Locatelli, M. Barisic, J. Andersen, Simon Bekker-Jensen, A. Lund, V. Rogov, Elena Papaleo, L. Lanzetti, Daniela Zio, F. Cecconi (2018)
Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellitesNature Communications, 10
( Ljubimov V , Ramesh A , Davani S , Danielpour M , Breunig J , Black K . Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID‐19 pandemic. Adv Drug Deliv Rev. 2022;181:114033.34808227)
Ljubimov V , Ramesh A , Davani S , Danielpour M , Breunig J , Black K . Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID‐19 pandemic. Adv Drug Deliv Rev. 2022;181:114033.34808227Ljubimov V , Ramesh A , Davani S , Danielpour M , Breunig J , Black K . Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID‐19 pandemic. Adv Drug Deliv Rev. 2022;181:114033.34808227, Ljubimov V , Ramesh A , Davani S , Danielpour M , Breunig J , Black K . Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID‐19 pandemic. Adv Drug Deliv Rev. 2022;181:114033.34808227
F. Iwamoto, L. Abrey, K. Beal, P. Gutin, M. Rosenblum, V. Reuter, L. DeAngelis, A. Lassman (2009)
Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastomaNeurology, 73
Juyeun Lee, Matthew Grabowski, J. Lathia (2022)
Tumor Treating Fields: killing two birds with one stoneThe Journal of Clinical Investigation, 132
P. Shi, Jia Tian, B. Ulm, Julianne Mallinger, H. Khoshbouei, L. Deleyrolle, M. Sarkisian (2021)
Tumor Treating Fields Suppression of Ciliogenesis Enhances Temozolomide ToxicityFrontiers in Oncology, 12
Eun Kim, W. Lee, Hoon-Kyu Oh (2021)
Tumor-treating fields in combination with sorafenib curtails the growth of colorectal carcinoma by inactivating AKT/STAT3 signalingTranslational Cancer Research, 11
V. Ljubimov, Arshia Ramesh, Saya Davani, M. Danielpour, Joshua Breunig, K. Black (2021)
Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID-19 pandemicAdvanced Drug Delivery Reviews, 181
D. Brandsma, L. Stalpers, W. Taal, P. Sminia, M. Bent (2008)
Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas.The Lancet. Oncology, 9 5
( Neuhaus E , Zirjacks L , Ganser K , et al. Alternating electric fields (TTFields) activate Ca1.2 channels in human glioblastoma cells. Cancer. 2019;11(1):110.)
Neuhaus E , Zirjacks L , Ganser K , et al. Alternating electric fields (TTFields) activate Ca1.2 channels in human glioblastoma cells. Cancer. 2019;11(1):110.Neuhaus E , Zirjacks L , Ganser K , et al. Alternating electric fields (TTFields) activate Ca1.2 channels in human glioblastoma cells. Cancer. 2019;11(1):110., Neuhaus E , Zirjacks L , Ganser K , et al. Alternating electric fields (TTFields) activate Ca1.2 channels in human glioblastoma cells. Cancer. 2019;11(1):110.
( Lee J , Grabowski M , Lathia J . Tumor treating fields: killing two birds with one stone. J Clin Invest. 2022;132(8):e159073.)
Lee J , Grabowski M , Lathia J . Tumor treating fields: killing two birds with one stone. J Clin Invest. 2022;132(8):e159073.Lee J , Grabowski M , Lathia J . Tumor treating fields: killing two birds with one stone. J Clin Invest. 2022;132(8):e159073., Lee J , Grabowski M , Lathia J . Tumor treating fields: killing two birds with one stone. J Clin Invest. 2022;132(8):e159073.
A. Shteingauz, Y. Porat, T. Voloshin, R. Schneiderman, M. Munster, E. Zeevi, N. Kaynan, K. Gotlib, M. Giladi, E. Kirson, U. Weinberg, A. Kinzel, Y. Palti (2018)
AMPK-dependent autophagy upregulation serves as a survival mechanism in response to Tumor Treating Fields (TTFields)Cell Death & Disease, 9
( Taphoorn M , Dirven L , Kanner A , et al. Influence of treatment with tumor‐treating fields on health‐related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495‐504.29392280)
Taphoorn M , Dirven L , Kanner A , et al. Influence of treatment with tumor‐treating fields on health‐related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495‐504.29392280Taphoorn M , Dirven L , Kanner A , et al. Influence of treatment with tumor‐treating fields on health‐related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495‐504.29392280, Taphoorn M , Dirven L , Kanner A , et al. Influence of treatment with tumor‐treating fields on health‐related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495‐504.29392280
Jacob Fisher, D. Adamson (2021)
Current FDA-Approved Therapies for High-Grade Malignant GliomasBiomedicines, 9
Guoqiang Sun, D. Rong, Zhouxiao Li, Guangshun Sun, Fan Wu, Xiao Li, Hongyong Cao, Ye Cheng, Weiwei Tang, Yangbai Sun (2021)
Role of Small Molecule Targeted Compounds in Cancer: Progress, Opportunities, and ChallengesFrontiers in Cell and Developmental Biology, 9
( Aguilar AA , Ho MC , Chang E , et al. Permeabilizing cell membranes with electric fields. Cancers (Basel). 2021;13(9):2283.)
Aguilar AA , Ho MC , Chang E , et al. Permeabilizing cell membranes with electric fields. Cancers (Basel). 2021;13(9):2283.Aguilar AA , Ho MC , Chang E , et al. Permeabilizing cell membranes with electric fields. Cancers (Basel). 2021;13(9):2283., Aguilar AA , Ho MC , Chang E , et al. Permeabilizing cell membranes with electric fields. Cancers (Basel). 2021;13(9):2283.
Ke Xu, N. Yin, Min Peng, Efstathios Stamatiades, S. Chhangawala, Amy Shyu, Peng Li, Xian Zhang, Mytrang Do, Kristelle Capistrano, Chun Chou, C. Leslie, Ming Li (2021)
Glycolytic ATP fuels phosphoinositide 3-kinase signaling to support effector T helper 17 cell responses.Immunity, 54 5
Bo Han, Xiangqi Meng, Pengfei Wu, Ziwei Li, Siyi Li, Yangong Zhang, Caijun Zha, Qile Ye, Chuanlu Jiang, Jinquan Cai, Tao Jiang (2020)
ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in gliomaTheranostics, 10
C. Wenger, P. Miranda, R. Salvador, A. Thielscher, Z. Bomzon, M. Giladi, M. Mrugala, A. Korshoej (2018)
A Review on Tumor-Treating Fields (TTFields): Clinical Implications Inferred From Computational ModelingIEEE Reviews in Biomedical Engineering, 11
A. Kessler, Ellaine Salvador, D. Domröse, M. Burek, C. Schaeffer, C. Brami, T. Voloshin, M. Giladi, R. Ernestus, M. Löhr, C. Förster, C. Hagemann (2019)
Blood Brain Barrier (BBB) Integrity Is Affected By Tumor Treating Fields (TTFields) in Vitro and In VivoInternational Journal of Radiation Oncology*Biology*Physics
( Xu K , Yin N , Peng M , et al. Glycolytic ATP fuels phosphoinositide 3‐kinase signaling to support effector T helper 17 cell responses. Immunity. 2021;54(5):976‐987.33979589)
Xu K , Yin N , Peng M , et al. Glycolytic ATP fuels phosphoinositide 3‐kinase signaling to support effector T helper 17 cell responses. Immunity. 2021;54(5):976‐987.33979589Xu K , Yin N , Peng M , et al. Glycolytic ATP fuels phosphoinositide 3‐kinase signaling to support effector T helper 17 cell responses. Immunity. 2021;54(5):976‐987.33979589, Xu K , Yin N , Peng M , et al. Glycolytic ATP fuels phosphoinositide 3‐kinase signaling to support effector T helper 17 cell responses. Immunity. 2021;54(5):976‐987.33979589
( Zhao W , Steinfeld J , Liang F , et al. BRCA1‐BARD1 promotes RAD51‐mediated homologous DNA pairing. Nature. 2017;550(7676):360‐365.28976962)
Zhao W , Steinfeld J , Liang F , et al. BRCA1‐BARD1 promotes RAD51‐mediated homologous DNA pairing. Nature. 2017;550(7676):360‐365.28976962Zhao W , Steinfeld J , Liang F , et al. BRCA1‐BARD1 promotes RAD51‐mediated homologous DNA pairing. Nature. 2017;550(7676):360‐365.28976962, Zhao W , Steinfeld J , Liang F , et al. BRCA1‐BARD1 promotes RAD51‐mediated homologous DNA pairing. Nature. 2017;550(7676):360‐365.28976962
( Silginer M , Weller M , Stupp R , Roth P . Biological activity of tumor‐treating fields in preclinical glioma models. Cell Death Dis. 2017;8(4):e2753.28425987)
Silginer M , Weller M , Stupp R , Roth P . Biological activity of tumor‐treating fields in preclinical glioma models. Cell Death Dis. 2017;8(4):e2753.28425987Silginer M , Weller M , Stupp R , Roth P . Biological activity of tumor‐treating fields in preclinical glioma models. Cell Death Dis. 2017;8(4):e2753.28425987, Silginer M , Weller M , Stupp R , Roth P . Biological activity of tumor‐treating fields in preclinical glioma models. Cell Death Dis. 2017;8(4):e2753.28425987
( Langen K , Watts C . Neuro‐oncology: amino acid PET for brain tumours – ready for the clinic? Nat Rev Neurol. 2016;12(7):375‐376.27282652)
Langen K , Watts C . Neuro‐oncology: amino acid PET for brain tumours – ready for the clinic? Nat Rev Neurol. 2016;12(7):375‐376.27282652Langen K , Watts C . Neuro‐oncology: amino acid PET for brain tumours – ready for the clinic? Nat Rev Neurol. 2016;12(7):375‐376.27282652, Langen K , Watts C . Neuro‐oncology: amino acid PET for brain tumours – ready for the clinic? Nat Rev Neurol. 2016;12(7):375‐376.27282652
N. Mikic, F. Poulsen, K. Kristoffersen, R. Laursen, T. Guldberg, J. Skjøth-Rasmussen, E. Wong, S. Møller, R. Dahlrot, J. Sørensen, A. Korshøj (2021)
Study protocol for OptimalTTF-2: enhancing Tumor Treating Fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi-center, randomized, prospective, interventional trialBMC Cancer, 21
S. Toms, C. Kim, G. Nicholas, Z. Ram (2018)
Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF-14 phase III trialJournal of Neuro-Oncology, 141
( Barreca M , Stathis A , Barraja P , Bertoni F . An overview on anti‐tubulin agents for the treatment of lymphoma patients. Pharmacol Ther. 2020;211:107552.32305312)
Barreca M , Stathis A , Barraja P , Bertoni F . An overview on anti‐tubulin agents for the treatment of lymphoma patients. Pharmacol Ther. 2020;211:107552.32305312Barreca M , Stathis A , Barraja P , Bertoni F . An overview on anti‐tubulin agents for the treatment of lymphoma patients. Pharmacol Ther. 2020;211:107552.32305312, Barreca M , Stathis A , Barraja P , Bertoni F . An overview on anti‐tubulin agents for the treatment of lymphoma patients. Pharmacol Ther. 2020;211:107552.32305312
( Voloshin T , Kaynan N , Davidi S , et al. Tumor‐treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti‐PD‐1 therapy. Cancer Immunol Immunother: CII. 2020;69(7):1191‐1204.32144446)
Voloshin T , Kaynan N , Davidi S , et al. Tumor‐treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti‐PD‐1 therapy. Cancer Immunol Immunother: CII. 2020;69(7):1191‐1204.32144446Voloshin T , Kaynan N , Davidi S , et al. Tumor‐treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti‐PD‐1 therapy. Cancer Immunol Immunother: CII. 2020;69(7):1191‐1204.32144446, Voloshin T , Kaynan N , Davidi S , et al. Tumor‐treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti‐PD‐1 therapy. Cancer Immunol Immunother: CII. 2020;69(7):1191‐1204.32144446
( Iwamoto F , Abrey L , Beal K , et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200‐1206.19822869)
Iwamoto F , Abrey L , Beal K , et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200‐1206.19822869Iwamoto F , Abrey L , Beal K , et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200‐1206.19822869, Iwamoto F , Abrey L , Beal K , et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200‐1206.19822869
( Li X , Yang F , Rubinsky B . A theoretical study on the biophysical mechanisms by which tumor treating fields affect tumor cells during mitosis. IEEE Trans Biomed Eng. 2020;67(9):2594‐2602.31940516)
Li X , Yang F , Rubinsky B . A theoretical study on the biophysical mechanisms by which tumor treating fields affect tumor cells during mitosis. IEEE Trans Biomed Eng. 2020;67(9):2594‐2602.31940516Li X , Yang F , Rubinsky B . A theoretical study on the biophysical mechanisms by which tumor treating fields affect tumor cells during mitosis. IEEE Trans Biomed Eng. 2020;67(9):2594‐2602.31940516, Li X , Yang F , Rubinsky B . A theoretical study on the biophysical mechanisms by which tumor treating fields affect tumor cells during mitosis. IEEE Trans Biomed Eng. 2020;67(9):2594‐2602.31940516
( Brandsma D , Stalpers L , Taal W , Sminia P , van den Bent M . Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453‐461.18452856)
Brandsma D , Stalpers L , Taal W , Sminia P , van den Bent M . Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453‐461.18452856Brandsma D , Stalpers L , Taal W , Sminia P , van den Bent M . Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453‐461.18452856, Brandsma D , Stalpers L , Taal W , Sminia P , van den Bent M . Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453‐461.18452856
G. Chinigò, H. Castel, O. Chever, D. Gkika (2021)
TRP Channels in Brain TumorsFrontiers in Cell and Developmental Biology, 9
( Payne SL , Ram P , Srinivasan DH , Le TT , Levin M , Oudin MJ . Potassium channel‐driven bioelectric signalling regulates metastasis in triple‐negative breast cancer. EBioMedicine. 2022;75:103767.34933180)
Payne SL , Ram P , Srinivasan DH , Le TT , Levin M , Oudin MJ . Potassium channel‐driven bioelectric signalling regulates metastasis in triple‐negative breast cancer. EBioMedicine. 2022;75:103767.34933180Payne SL , Ram P , Srinivasan DH , Le TT , Levin M , Oudin MJ . Potassium channel‐driven bioelectric signalling regulates metastasis in triple‐negative breast cancer. EBioMedicine. 2022;75:103767.34933180, Payne SL , Ram P , Srinivasan DH , Le TT , Levin M , Oudin MJ . Potassium channel‐driven bioelectric signalling regulates metastasis in triple‐negative breast cancer. EBioMedicine. 2022;75:103767.34933180
( Kalluri R , Weinberg R . The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119(6):1420‐1428.19487818)
Kalluri R , Weinberg R . The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119(6):1420‐1428.19487818Kalluri R , Weinberg R . The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119(6):1420‐1428.19487818, Kalluri R , Weinberg R . The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119(6):1420‐1428.19487818
( Stupp R , Taillibert S , Kanner A , et al. Effect of tumor‐treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318(23):2306‐2316.29260225)
Stupp R , Taillibert S , Kanner A , et al. Effect of tumor‐treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318(23):2306‐2316.29260225Stupp R , Taillibert S , Kanner A , et al. Effect of tumor‐treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318(23):2306‐2316.29260225, Stupp R , Taillibert S , Kanner A , et al. Effect of tumor‐treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318(23):2306‐2316.29260225
M. Salzberg, E. Kirson, Y. Palti, C. Rochlitz (2008)
A Pilot Study with Very Low-Intensity, Intermediate-Frequency Electric Fields in Patients with Locally Advanced and/or Metastatic Solid TumorsOncology Research and Treatment, 31
( Bokstein F , Blumenthal D , Limon D , Harosh C , Ram Z , Grossman R . Concurrent tumor treating fields (TTFields) and radiation therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411.32373508)
Bokstein F , Blumenthal D , Limon D , Harosh C , Ram Z , Grossman R . Concurrent tumor treating fields (TTFields) and radiation therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411.32373508Bokstein F , Blumenthal D , Limon D , Harosh C , Ram Z , Grossman R . Concurrent tumor treating fields (TTFields) and radiation therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411.32373508, Bokstein F , Blumenthal D , Limon D , Harosh C , Ram Z , Grossman R . Concurrent tumor treating fields (TTFields) and radiation therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411.32373508
( Chinigò G , Castel H , Chever O , Gkika D . TRP channels in brain tumors. Front Cell Dev Biol. 2021;9:617801.33928077)
Chinigò G , Castel H , Chever O , Gkika D . TRP channels in brain tumors. Front Cell Dev Biol. 2021;9:617801.33928077Chinigò G , Castel H , Chever O , Gkika D . TRP channels in brain tumors. Front Cell Dev Biol. 2021;9:617801.33928077, Chinigò G , Castel H , Chever O , Gkika D . TRP channels in brain tumors. Front Cell Dev Biol. 2021;9:617801.33928077
( DeBerardinis R , Chandel N . We need to talk about the Warburg effect. Nat Metab. 2020;2(2):127‐129.32694689)
DeBerardinis R , Chandel N . We need to talk about the Warburg effect. Nat Metab. 2020;2(2):127‐129.32694689DeBerardinis R , Chandel N . We need to talk about the Warburg effect. Nat Metab. 2020;2(2):127‐129.32694689, DeBerardinis R , Chandel N . We need to talk about the Warburg effect. Nat Metab. 2020;2(2):127‐129.32694689
( Patel C , Beinat C , Xie Y , Chang E , Gambhir S . Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [F]DASA‐23, a non‐invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia (New York, NY). 2021;23(1):58‐67.)
Patel C , Beinat C , Xie Y , Chang E , Gambhir S . Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [F]DASA‐23, a non‐invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia (New York, NY). 2021;23(1):58‐67.Patel C , Beinat C , Xie Y , Chang E , Gambhir S . Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [F]DASA‐23, a non‐invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia (New York, NY). 2021;23(1):58‐67., Patel C , Beinat C , Xie Y , Chang E , Gambhir S . Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [F]DASA‐23, a non‐invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia (New York, NY). 2021;23(1):58‐67.
( Probst U , Fuhrmann I , Beyer L , Wiggermann P . Electrochemotherapy as a new modality in interventional oncology: a review. Technol Cancer Res Treat. 2018;17:1533033818785329.)
Probst U , Fuhrmann I , Beyer L , Wiggermann P . Electrochemotherapy as a new modality in interventional oncology: a review. Technol Cancer Res Treat. 2018;17:1533033818785329.Probst U , Fuhrmann I , Beyer L , Wiggermann P . Electrochemotherapy as a new modality in interventional oncology: a review. Technol Cancer Res Treat. 2018;17:1533033818785329., Probst U , Fuhrmann I , Beyer L , Wiggermann P . Electrochemotherapy as a new modality in interventional oncology: a review. Technol Cancer Res Treat. 2018;17:1533033818785329.
A. Achrol, R. Rennert, C. Anders, R. Soffietti, M. Ahluwalia, L. Nayak, S. Peters, Nils Arvold, G. Harsh, P. Steeg, Steven Chang (2019)
Brain metastasesNature Reviews Disease Primers, 5
T. Singhal, T. Narayanan, V. Jain, J. Mukherjee, J. Mantil (2007)
11C-l-Methionine Positron Emission Tomography in the Clinical Management of Cerebral GliomasMolecular Imaging and Biology, 10
( Mehta M , Wen P , Nishikawa R , Reardon D , Peters K . Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60‐65.28259296)
Mehta M , Wen P , Nishikawa R , Reardon D , Peters K . Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60‐65.28259296Mehta M , Wen P , Nishikawa R , Reardon D , Peters K . Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60‐65.28259296, Mehta M , Wen P , Nishikawa R , Reardon D , Peters K . Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60‐65.28259296
R. Lukas, P. Gabikian, M. Garza, S. Chmura (2014)
Treatment of Brain MetastasesOncology, 87
Alondra Aguilar, Michelle Ho, E. Chang, Kristen Carlson, A. Natarajan, T. Marciano, Z. Bomzon, Chirag Patel (2021)
Permeabilizing Cell Membranes with Electric FieldsCancers, 13
( Den R , Kamrava M , Sheng Z , et al. A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high‐grade gliomas. Int J Radiat Oncol Biol Phys. 2013;85(2):321‐328.22687197)
Den R , Kamrava M , Sheng Z , et al. A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high‐grade gliomas. Int J Radiat Oncol Biol Phys. 2013;85(2):321‐328.22687197Den R , Kamrava M , Sheng Z , et al. A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high‐grade gliomas. Int J Radiat Oncol Biol Phys. 2013;85(2):321‐328.22687197, Den R , Kamrava M , Sheng Z , et al. A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high‐grade gliomas. Int J Radiat Oncol Biol Phys. 2013;85(2):321‐328.22687197
K. Langen, C. Watts (2016)
Neuro-oncology: Amino acid PET for brain tumours — ready for the clinic?Nature Reviews Neurology, 12
( Singhal T , Narayanan T , Jain V , Mukherjee J , Mantil J . 11C‐L‐methionine positron emission tomography in the clinical management of cerebral gliomas. Mol Imaging Biol. 2008;10(1):1‐18.17957408)
Singhal T , Narayanan T , Jain V , Mukherjee J , Mantil J . 11C‐L‐methionine positron emission tomography in the clinical management of cerebral gliomas. Mol Imaging Biol. 2008;10(1):1‐18.17957408Singhal T , Narayanan T , Jain V , Mukherjee J , Mantil J . 11C‐L‐methionine positron emission tomography in the clinical management of cerebral gliomas. Mol Imaging Biol. 2008;10(1):1‐18.17957408, Singhal T , Narayanan T , Jain V , Mukherjee J , Mantil J . 11C‐L‐methionine positron emission tomography in the clinical management of cerebral gliomas. Mol Imaging Biol. 2008;10(1):1‐18.17957408
( Toms S , Kim C , Nicholas G , Ram Z . Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF‐14 phase III trial. J Neurooncol. 2019;141(2):467‐473.30506499)
Toms S , Kim C , Nicholas G , Ram Z . Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF‐14 phase III trial. J Neurooncol. 2019;141(2):467‐473.30506499Toms S , Kim C , Nicholas G , Ram Z . Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF‐14 phase III trial. J Neurooncol. 2019;141(2):467‐473.30506499, Toms S , Kim C , Nicholas G , Ram Z . Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF‐14 phase III trial. J Neurooncol. 2019;141(2):467‐473.30506499
( Gilden J , Peck S , Chen Y , Krummel M . The septin cytoskeleton facilitates membrane retraction during motility and blebbing. J Cell Biol. 2012;196(1):103‐114.22232702)
Gilden J , Peck S , Chen Y , Krummel M . The septin cytoskeleton facilitates membrane retraction during motility and blebbing. J Cell Biol. 2012;196(1):103‐114.22232702Gilden J , Peck S , Chen Y , Krummel M . The septin cytoskeleton facilitates membrane retraction during motility and blebbing. J Cell Biol. 2012;196(1):103‐114.22232702, Gilden J , Peck S , Chen Y , Krummel M . The septin cytoskeleton facilitates membrane retraction during motility and blebbing. J Cell Biol. 2012;196(1):103‐114.22232702
N. Gatson, J. Barnholtz-Sloan, J. Drappatz, R. Henriksson, A. Hottinger, P. Hinoul, C. Kruchko, V. Puduvalli, D. Tran, E. Wong, M. Glas (2021)
Tumor Treating Fields for Glioblastoma Therapy During the COVID-19 PandemicFrontiers in Oncology, 11
F. Bernard-Arnoux, M. Lamure, F. Ducray, Gilles Aulagner, J. Honnorat, Xavier Armoiry (2016)
The cost-effectiveness of tumor-treating fields therapy in patients with newly diagnosed glioblastoma.Neuro-oncology, 18 8
R. Rivoirard, A. Vallard, J. Langrand-Escure, M. Mrad, Guoping Wang, J. Guy, P. Diao, Alexandre Dubanchet, E. Deutsch, C. Rancoule, N. Magné (2016)
Thirty years of phase I radiochemotherapy trials: Latest development.European journal of cancer, 58
( Witney T , James M , Shen B , et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med. 2015;7(310):310ra169.)
Witney T , James M , Shen B , et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med. 2015;7(310):310ra169.Witney T , James M , Shen B , et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med. 2015;7(310):310ra169., Witney T , James M , Shen B , et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med. 2015;7(310):310ra169.
( Kessler A , Frömbling G , Gross F , et al. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov. 2018;4:12.)
Kessler A , Frömbling G , Gross F , et al. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov. 2018;4:12.Kessler A , Frömbling G , Gross F , et al. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov. 2018;4:12., Kessler A , Frömbling G , Gross F , et al. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov. 2018;4:12.
Xing Li, Fan Yang, B. Rubinsky (2020)
A Theoretical Study on the Biophysical Mechanisms by Which Tumor Treating Fields Affect Tumor Cells During MitosisIEEE Transactions on Biomedical Engineering, 67
( Sun G , Rong D , Li Z , et al. Role of small molecule targeted compounds in cancer: Progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363.34568317)
Sun G , Rong D , Li Z , et al. Role of small molecule targeted compounds in cancer: Progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363.34568317Sun G , Rong D , Li Z , et al. Role of small molecule targeted compounds in cancer: Progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363.34568317, Sun G , Rong D , Li Z , et al. Role of small molecule targeted compounds in cancer: Progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363.34568317
Zeinab Akbarnejad, H. Eskandary, L. Dini, C. Vergallo, S. Nematollahi-Mahani, A. Farsinejad, M. Abadi, Meysam Ahmadi (2017)
Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low-frequency electromagnetic field (100Hz, 100G).Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie, 92
( Wenger C , Miranda P , Salvador R , et al. A review on tumor‐treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195‐207.29993870)
Wenger C , Miranda P , Salvador R , et al. A review on tumor‐treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195‐207.29993870Wenger C , Miranda P , Salvador R , et al. A review on tumor‐treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195‐207.29993870, Wenger C , Miranda P , Salvador R , et al. A review on tumor‐treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195‐207.29993870
( Tian M , He X , Jin C , et al. Transpathology: molecular imaging‐based pathology. Eur J Nucl Med Mol Imaging. 2021;48(8):2338‐2350.33585964)
Tian M , He X , Jin C , et al. Transpathology: molecular imaging‐based pathology. Eur J Nucl Med Mol Imaging. 2021;48(8):2338‐2350.33585964Tian M , He X , Jin C , et al. Transpathology: molecular imaging‐based pathology. Eur J Nucl Med Mol Imaging. 2021;48(8):2338‐2350.33585964, Tian M , He X , Jin C , et al. Transpathology: molecular imaging‐based pathology. Eur J Nucl Med Mol Imaging. 2021;48(8):2338‐2350.33585964
M. Majores, M. Lehe, J. Fassunke, J. Schramm, A. Becker, M. Simon (2008)
Tumor recurrence and malignant progression of gangliogliomasCancer, 113
S. Lee, Adrian Tunkyi, E. Wong, K. Swanson (2012)
Mitosis interference of cancer cells during anaphase by electric field from NovoTTF-100A: An update.Journal of Clinical Oncology, 30
Julia Gilden, Sebastian Peck, Yi-Chun Chen, M. Krummel (2012)
The septin cytoskeleton facilitates membrane retraction during motility and blebbingThe Journal of Cell Biology, 196
(2010)
Modeling tumor growth kinetics and its implications for TTFields treatment planning
E. Neuhaus, Lisa Zirjacks, K. Ganser, L. Klumpp, Uwe Schüler, D. Zips, F. Eckert, S. Huber (2019)
Alternating Electric Fields (TTFields) Activate Cav1.2 Channels in Human Glioblastoma CellsCancers, 11
Zeina Kais, B. Rondinelli, A. Holmes, Colin O’Leary, D. Kozono, A. D’Andrea, Raphael Ceccaldi (2016)
FANCD2 Maintains Fork Stability in BRCA1/2-Deficient Tumors and Promotes Alternative End-Joining DNA Repair.Cell reports, 15 11
E. Kirson, Zoya Gurvich, R. Schneiderman, E. Dekel, A. Itzhaki, Yoram Wasserman, R. Schatzberger, Y. Palti (2004)
Disruption of cancer cell replication by alternating electric fields.Cancer research, 64 9
( Arvanitis CD , Ferraro GB , Jain RK . The blood‐brain barrier and blood‐tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41.31601988)
Arvanitis CD , Ferraro GB , Jain RK . The blood‐brain barrier and blood‐tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41.31601988Arvanitis CD , Ferraro GB , Jain RK . The blood‐brain barrier and blood‐tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41.31601988, Arvanitis CD , Ferraro GB , Jain RK . The blood‐brain barrier and blood‐tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41.31601988
( Davis M . Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420‐429.30392758)
Davis M . Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420‐429.30392758Davis M . Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420‐429.30392758, Davis M . Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420‐429.30392758
E. Bosnyák, G. Barger, S. Michelhaugh, N. Robinette, Alit Amit-Yousif, S. Mittal, C. Juhász (2017)
Amino Acid PET Imaging of the Early Metabolic Response During Tumor-Treating Fields (TTFields) Therapy in Recurrent Glioblastoma.Clinical Nuclear Medicine
( Palti Y . Stimulation of muscles and nerves by means of externally applied electrodes. Bull Res Counc Isr Sect E Exp Med. 1962;10:54‐56.)
Palti Y . Stimulation of muscles and nerves by means of externally applied electrodes. Bull Res Counc Isr Sect E Exp Med. 1962;10:54‐56.Palti Y . Stimulation of muscles and nerves by means of externally applied electrodes. Bull Res Counc Isr Sect E Exp Med. 1962;10:54‐56., Palti Y . Stimulation of muscles and nerves by means of externally applied electrodes. Bull Res Counc Isr Sect E Exp Med. 1962;10:54‐56.
( Levin M , Stevenson CG . Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering. Annu Rev Biomed Eng. 2012;14(1):295‐323.22809139)
Levin M , Stevenson CG . Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering. Annu Rev Biomed Eng. 2012;14(1):295‐323.22809139Levin M , Stevenson CG . Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering. Annu Rev Biomed Eng. 2012;14(1):295‐323.22809139, Levin M , Stevenson CG . Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering. Annu Rev Biomed Eng. 2012;14(1):295‐323.22809139
J. Rosen, T. Blau, S. Grau, M. Barbe, G. Fink, N. Galldiks (2018)
Extracranial Metastases of a Cerebral Glioblastoma: A Case Report and Review of the LiteratureCase Reports in Oncology, 11
W. Wick (2016)
TTFields: where does all the skepticism come from?Neuro-oncology, 18 3
( Green A , Mulcahy Levy J , Vibhakar R , et al. Tumor treating fields in pediatric high‐grade glioma. Child's Nerv Syst: ChNS: Off J Int Soc Pediat Neurosurg. 2017;33(7):1043‐1045.)
Green A , Mulcahy Levy J , Vibhakar R , et al. Tumor treating fields in pediatric high‐grade glioma. Child's Nerv Syst: ChNS: Off J Int Soc Pediat Neurosurg. 2017;33(7):1043‐1045.Green A , Mulcahy Levy J , Vibhakar R , et al. Tumor treating fields in pediatric high‐grade glioma. Child's Nerv Syst: ChNS: Off J Int Soc Pediat Neurosurg. 2017;33(7):1043‐1045., Green A , Mulcahy Levy J , Vibhakar R , et al. Tumor treating fields in pediatric high‐grade glioma. Child's Nerv Syst: ChNS: Off J Int Soc Pediat Neurosurg. 2017;33(7):1043‐1045.
M. Taphoorn, L. Dirven, A. Kanner, G. Lavy-Shahaf, U. Weinberg, S. Taillibert, S. Toms, J. Honnorat, Thomas Chen, J. Šroubek, C. David, A. Idbaih, J. Easaw, Chae-Yong Kim, J. Bruna, A. Hottinger, Y. Kew, P. Roth, R. Desai, J. Villano, E. Kirson, Z. Ram, R. Stupp (2018)
Influence of Treatment With Tumor-Treating Fields on Health-Related Quality of Life of Patients With Newly Diagnosed Glioblastoma: A Secondary Analysis of a Randomized Clinical TrialJAMA Oncology, 4
( Majores M , von Lehe M , Fassunke J , Schramm J , Becker AJ , Simon M . Tumor recurrence and malignant progression of gangliogliomas. Cancer. 2008;113(12):3355‐3363.18988291)
Majores M , von Lehe M , Fassunke J , Schramm J , Becker AJ , Simon M . Tumor recurrence and malignant progression of gangliogliomas. Cancer. 2008;113(12):3355‐3363.18988291Majores M , von Lehe M , Fassunke J , Schramm J , Becker AJ , Simon M . Tumor recurrence and malignant progression of gangliogliomas. Cancer. 2008;113(12):3355‐3363.18988291, Majores M , von Lehe M , Fassunke J , Schramm J , Becker AJ , Simon M . Tumor recurrence and malignant progression of gangliogliomas. Cancer. 2008;113(12):3355‐3363.18988291
S. Mohan, S. Chawla, Sumei Wang, G. Verma, A. Skolnik, S. Brem, K. Peters, Harish Poptani (2016)
Assessment of early response to tumor-treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience.CNS oncology, 5 3
Eun Kim, Hyo-Sook Song, S. Yoo, M. Yoon (2016)
Tumor treating fields inhibit glioblastoma cell migration, invasion and angiogenesisOncotarget, 7
L. Siems, A. Kosman, S. Osborne (1948)
A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating.Archives of physical medicine and rehabilitation, 29 12
M. Giladi, R. Schneiderman, Y. Porat, M. Munster, A. Itzhaki, D. Mordechovich, S. Cahal, E. Kirson, U. Weinberg, Y. Palti (2014)
Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields.Pancreatology : official journal of the International Association of Pancreatology (IAP) ... [et al.], 14 1
Table II.
( Werner J , Lohmann P , Fink G , Langen K , Galldiks N . Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules (Basel, Switzerland). 2020;25(6):1471.)
Werner J , Lohmann P , Fink G , Langen K , Galldiks N . Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules (Basel, Switzerland). 2020;25(6):1471.Werner J , Lohmann P , Fink G , Langen K , Galldiks N . Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules (Basel, Switzerland). 2020;25(6):1471., Werner J , Lohmann P , Fink G , Langen K , Galldiks N . Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules (Basel, Switzerland). 2020;25(6):1471.
H. Christofk, M. Heiden, M. Harris, A. Ramanathan, R. Gerszten, Ru Wei, M. Fleming, S. Schreiber, L. Cantley (2008)
The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growthNature, 452
M. Mrugala (2009)
Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence.Neurology, 72 8
( Shteingauz A , Porat Y , Voloshin T , et al. AMPK‐dependent autophagy upregulation serves as a survival mechanism in response to tumor treating fields (TTFields). Cell Death Dis. 2018;9(11):1074.30341282)
Shteingauz A , Porat Y , Voloshin T , et al. AMPK‐dependent autophagy upregulation serves as a survival mechanism in response to tumor treating fields (TTFields). Cell Death Dis. 2018;9(11):1074.30341282Shteingauz A , Porat Y , Voloshin T , et al. AMPK‐dependent autophagy upregulation serves as a survival mechanism in response to tumor treating fields (TTFields). Cell Death Dis. 2018;9(11):1074.30341282, Shteingauz A , Porat Y , Voloshin T , et al. AMPK‐dependent autophagy upregulation serves as a survival mechanism in response to tumor treating fields (TTFields). Cell Death Dis. 2018;9(11):1074.30341282
E. Jackson, D. Barboriak, L. Bidaut, C. Meyer (2009)
Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources.Translational oncology, 2 4
M. Giladi, M. Munster, R. Schneiderman, T. Voloshin, Y. Porat, R. Blat, Katarzyna Zielinska-Chomej, P. Hååg, Z. Bomzon, E. Kirson, U. Weinberg, K. Viktorsson, R. Lewensohn, Y. Palti (2017)
Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cellsRadiation Oncology (London, England), 12
( Kirson ED , Wasserman Y , Izhaki A , et al. Modeling tumor growth kinetics and its implications for TTFields treatment planning. Neuro Oncol. 2010;12:48.)
Kirson ED , Wasserman Y , Izhaki A , et al. Modeling tumor growth kinetics and its implications for TTFields treatment planning. Neuro Oncol. 2010;12:48.Kirson ED , Wasserman Y , Izhaki A , et al. Modeling tumor growth kinetics and its implications for TTFields treatment planning. Neuro Oncol. 2010;12:48., Kirson ED , Wasserman Y , Izhaki A , et al. Modeling tumor growth kinetics and its implications for TTFields treatment planning. Neuro Oncol. 2010;12:48.
( Giladi M , Schneiderman R , Porat Y , et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatol: Off J Int Assoc Pancreatol (IAP). 2014;14(1):54‐63.)
Giladi M , Schneiderman R , Porat Y , et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatol: Off J Int Assoc Pancreatol (IAP). 2014;14(1):54‐63.Giladi M , Schneiderman R , Porat Y , et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatol: Off J Int Assoc Pancreatol (IAP). 2014;14(1):54‐63., Giladi M , Schneiderman R , Porat Y , et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatol: Off J Int Assoc Pancreatol (IAP). 2014;14(1):54‐63.
silviya Meletath, D. Pavlick, Tim Brennan, R. Hamilton, J. Chmielecki, J. Elvin, N. Palma, J. Ross, V. Miller, P. Stephens, G. Snipes, V. Rajaram, Siraj Ali, I. Melguizo-Gavilanes (2016)
Personalized Treatment for a Patient With a BRAF V600E Mutation Using Dabrafenib and a Tumor Treatment Fields Device in a High-Grade Glioma Arising From Ganglioglioma.Journal of the National Comprehensive Cancer Network : JNCCN, 14 11
Shengchao Xu, C. Luo, Dikang Chen, Lu Tang, Ling Chen, Zhixiong Liu (2022)
Whole transcriptome and proteome analyses identify potential targets and mechanisms underlying tumor treating fields against glioblastomaCell Death & Disease, 13
Jeong-In Park, K. Song, Seung-Youn Jung, J. Ahn, Sang-Gu Hwang, Joon Kim, Eun Kim, Jie-Young Song (2019)
Tumor-Treating Fields Induce RAW264.7 Macrophage Activation Via NK-κB/MAPK Signaling PathwaysTechnology in Cancer Research & Treatment, 18
M. Silginer, M. Weller, R. Stupp, P. Roth (2017)
Biological activity of tumor-treating fields in preclinical glioma modelsCell Death & Disease, 8
( Lu G , Rao M , Zhu P , et al. Triple‐drug therapy with bevacizumab, irinotecan, and temozolomide plus tumor treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.30766509)
Lu G , Rao M , Zhu P , et al. Triple‐drug therapy with bevacizumab, irinotecan, and temozolomide plus tumor treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.30766509Lu G , Rao M , Zhu P , et al. Triple‐drug therapy with bevacizumab, irinotecan, and temozolomide plus tumor treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.30766509, Lu G , Rao M , Zhu P , et al. Triple‐drug therapy with bevacizumab, irinotecan, and temozolomide plus tumor treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.30766509
( Vergote I , von Moos R , Manso L , Van Nieuwenhuysen E , Concin N , Sessa C . Tumor treating fields in combination with paclitaxel in recurrent ovarian carcinoma: results of the INNOVATE pilot study. Gynecol Oncol. 2018;150(3):471‐477.30060963)
Vergote I , von Moos R , Manso L , Van Nieuwenhuysen E , Concin N , Sessa C . Tumor treating fields in combination with paclitaxel in recurrent ovarian carcinoma: results of the INNOVATE pilot study. Gynecol Oncol. 2018;150(3):471‐477.30060963Vergote I , von Moos R , Manso L , Van Nieuwenhuysen E , Concin N , Sessa C . Tumor treating fields in combination with paclitaxel in recurrent ovarian carcinoma: results of the INNOVATE pilot study. Gynecol Oncol. 2018;150(3):471‐477.30060963, Vergote I , von Moos R , Manso L , Van Nieuwenhuysen E , Concin N , Sessa C . Tumor treating fields in combination with paclitaxel in recurrent ovarian carcinoma: results of the INNOVATE pilot study. Gynecol Oncol. 2018;150(3):471‐477.30060963
E. Kirson, M. Giladi, Zoya Gurvich, A. Itzhaki, D. Mordechovich, R. Schneiderman, Yoram Wasserman, B. Ryffel, D. Goldsher, Y. Palti (2009)
Alternating electric fields (TTFields) inhibit metastatic spread of solid tumors to the lungsClinical & Experimental Metastasis, 26
( Haj A , Doenitz C , Hohenberger C , Brawanski A , Proescholdt MA . P01.144 TTFields for newly diagnosed glioblastoma: impact of consultation strategy. (1522–8517 (Print)).)
Haj A , Doenitz C , Hohenberger C , Brawanski A , Proescholdt MA . P01.144 TTFields for newly diagnosed glioblastoma: impact of consultation strategy. (1522–8517 (Print)).Haj A , Doenitz C , Hohenberger C , Brawanski A , Proescholdt MA . P01.144 TTFields for newly diagnosed glioblastoma: impact of consultation strategy. (1522–8517 (Print))., Haj A , Doenitz C , Hohenberger C , Brawanski A , Proescholdt MA . P01.144 TTFields for newly diagnosed glioblastoma: impact of consultation strategy. (1522–8517 (Print)).
A. Robinson, Akhil Jain, Harry Sherman, R. Hague, R. Rahman, Paola Sanjuan‐Alberte, F. Rawson (2021)
Toward Hijacking Bioelectricity in Cancer to Develop New Bioelectronic MedicineAdvanced Therapeutics, 4
( Kirschenbaum D , Amit I . Brain metastases: not all tumors are created equal. Neuron. 2022;110(7):1097‐1099.35390289)
Kirschenbaum D , Amit I . Brain metastases: not all tumors are created equal. Neuron. 2022;110(7):1097‐1099.35390289Kirschenbaum D , Amit I . Brain metastases: not all tumors are created equal. Neuron. 2022;110(7):1097‐1099.35390289, Kirschenbaum D , Amit I . Brain metastases: not all tumors are created equal. Neuron. 2022;110(7):1097‐1099.35390289
( Inoue T , Nakayama Y , Li Y , et al. SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitors. FEBS J. 2014;281(11):2623‐2637.24712640)
Inoue T , Nakayama Y , Li Y , et al. SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitors. FEBS J. 2014;281(11):2623‐2637.24712640Inoue T , Nakayama Y , Li Y , et al. SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitors. FEBS J. 2014;281(11):2623‐2637.24712640, Inoue T , Nakayama Y , Li Y , et al. SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitors. FEBS J. 2014;281(11):2623‐2637.24712640
A. Norden, G. Young, K. Setayesh, A. Muzikansky, R. Klufas, G. Ross, A. Ciampa, L. Ebbeling, B. Levy, J. Drappatz, S. Kesari, P. Wen (2008)
Bevacizumab for recurrent malignant gliomasNeurology, 70
Samantha Payne, Priyanka Ram, D. Srinivasan, T. Le, M. Levin, Madeleine Oudin (2021)
Potassium channel-driven bioelectric signalling regulates metastasis in triple-negative breast cancerEBioMedicine, 75
T. Voloshin, N. Kaynan, S. Davidi, Y. Porat, A. Shteingauz, R. Schneiderman, E. Zeevi, M. Munster, R. Blat, C. Brami, S. Cahal, A. Itzhaki, M. Giladi, E. Kirson, U. Weinberg, A. Kinzel, Y. Palti (2020)
Tumor-treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti-PD-1 therapyCancer Immunology, Immunotherapy, 69
( Han B , Meng X , Wu P , et al. ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in glioma. Theranostics. 2020;10(7):3351‐3365.32194873)
Han B , Meng X , Wu P , et al. ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in glioma. Theranostics. 2020;10(7):3351‐3365.32194873Han B , Meng X , Wu P , et al. ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in glioma. Theranostics. 2020;10(7):3351‐3365.32194873, Han B , Meng X , Wu P , et al. ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in glioma. Theranostics. 2020;10(7):3351‐3365.32194873
Q. Ostrom, H. Gittleman, Gabrielle Truitt, Alexander Boscia, C. Kruchko, J. Barnholtz-Sloan (2018)
CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2011-2015.Neuro-oncology, 20 suppl_4
P. Gutin, E. Wong (2012)
Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality.American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting
R. Den, M. Kamrava, Z. Sheng, M. Werner-Wasik, E. Dougherty, Michelle Marinucchi, Y. Lawrence, S. Hegarty, T. Hyslop, D. Andrews, J. Glass, D. Friedman, Michael Green, K. Camphausen, A. Dicker (2013)
A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high-grade gliomas.International journal of radiation oncology, biology, physics, 85 2
E. Chang, C. Patel, C. Pohling, C. Young, Jonathan Song, T. Flores, Yitian Zeng, L. Joubert, H. Arami, A. Natarajan, R. Sinclair, S. Gambhir (2018)
Tumor treating fields increases membrane permeability in glioblastoma cellsCell Death Discovery, 4
( Gutin P , Wong E . Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality. American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Annual Meeting 2012:126–131.)
Gutin P , Wong E . Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality. American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Annual Meeting 2012:126–131.Gutin P , Wong E . Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality. American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Annual Meeting 2012:126–131., Gutin P , Wong E . Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality. American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Annual Meeting 2012:126–131.
R. Stupp, S. Taillibert, A. Kanner, W. Read, D. Steinberg, B. Lhermitte, S. Toms, A. Idbaih, M. Ahluwalia, K. Fink, F. Meco, F. Lieberman, Jay-Jiguang Zhu, G. Stragliotto, D. Tran, S. Brem, A. Hottinger, E. Kirson, G. Lavy-Shahaf, U. Weinberg, Chae-Yong Kim, S. Paek, G. Nicholas, Jordi Burna, H. Hirte, M. Weller, Y. Palti, M. Hegi, Z. Ram (2017)
Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical TrialJAMA, 318
M. Davis (2018)
Epidemiology and Overview of Gliomas.Seminars in oncology nursing, 34 5
( Gatson N , Barnholtz‐Sloan J , Drappatz J , et al. Tumor treating fields for glioblastoma therapy during the COVID‐19 pandemic. Front Oncol. 2021;11:679702.34026655)
Gatson N , Barnholtz‐Sloan J , Drappatz J , et al. Tumor treating fields for glioblastoma therapy during the COVID‐19 pandemic. Front Oncol. 2021;11:679702.34026655Gatson N , Barnholtz‐Sloan J , Drappatz J , et al. Tumor treating fields for glioblastoma therapy during the COVID‐19 pandemic. Front Oncol. 2021;11:679702.34026655, Gatson N , Barnholtz‐Sloan J , Drappatz J , et al. Tumor treating fields for glioblastoma therapy during the COVID‐19 pandemic. Front Oncol. 2021;11:679702.34026655
A. Kessler, Greta Frömbling, F. Gross, Mirja Hahn, Wilfrid Dzokou, R. Ernestus, M. Löhr, C. Hagemann (2018)
Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibitionCell Death Discovery, 4
T. Witney, M. James, Bin Shen, E. Chang, C. Pohling, Natasha Arksey, A. Hoehne, A. Shuhendler, Jun-Hyung Park, D. Bodapati, Judith Weber, G. Gowrishankar, J. Rao, F. Chin, S. Gambhir (2015)
PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2Science Translational Medicine, 7
Ellaine Salvador, Theresa Köppl, J. Hörmann, S. Schönhärl, Polina Bugaeva, A. Kessler, M. Burek, R. Ernestus, M. Löhr, C. Hagemann (2023)
Tumor Treating Fields (TTFields) Induce Cell Junction Alterations in a Human 3D In Vitro Model of the Blood-Brain BarrierPharmaceutics, 15
E. Wong, E. Lok, Sabrena Gautam, K. Swanson (2015)
Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastomaBritish Journal of Cancer, 113
( Siems LL , Kosman AJ , Osborne SL . A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating. Arch Phys Med Rehabil. 1948;29(12):759‐764.18107278)
Siems LL , Kosman AJ , Osborne SL . A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating. Arch Phys Med Rehabil. 1948;29(12):759‐764.18107278Siems LL , Kosman AJ , Osborne SL . A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating. Arch Phys Med Rehabil. 1948;29(12):759‐764.18107278, Siems LL , Kosman AJ , Osborne SL . A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating. Arch Phys Med Rehabil. 1948;29(12):759‐764.18107278
I. Martins, Y. Wang, Y. Wang, Y. Wang, M. Michaud, M. Michaud, M. Michaud, Yuting Ma, Yuting Ma, Yuting Ma, Abdul Sukkurwala, Abdul Sukkurwala, Abdul Sukkurwala, Shensi Shen, Shensi Shen, Shensi Shen, O. Kepp, O. Kepp, O. Kepp, D. Métivier, D. Métivier, D. Métivier, L. Galluzzi, L. Galluzzi, J-L Perfettini, J-L Perfettini, J-L Perfettini, L. Zitvogel, G. Kroemer (2013)
Molecular mechanisms of ATP secretion during immunogenic cell deathCell Death and Differentiation, 21
( Rosen J , Blau T , Grau S , Barbe M , Fink G , Galldiks N . Extracranial metastases of a cerebral glioblastoma: a case report and review of the literature. Case Reports Oncol. 2018;11(2):591‐600.)
Rosen J , Blau T , Grau S , Barbe M , Fink G , Galldiks N . Extracranial metastases of a cerebral glioblastoma: a case report and review of the literature. Case Reports Oncol. 2018;11(2):591‐600.Rosen J , Blau T , Grau S , Barbe M , Fink G , Galldiks N . Extracranial metastases of a cerebral glioblastoma: a case report and review of the literature. Case Reports Oncol. 2018;11(2):591‐600., Rosen J , Blau T , Grau S , Barbe M , Fink G , Galldiks N . Extracranial metastases of a cerebral glioblastoma: a case report and review of the literature. Case Reports Oncol. 2018;11(2):591‐600.
D. Kirschenbaum, I. Amit (2022)
Brain metastases: Not all tumors are created equalNeuron, 110
J. Werner, P. Lohmann, G. Fink, K. Langen, N. Galldiks (2020)
Current Landscape and Emerging Fields of PET Imaging in Patients with Brain TumorsMolecules, 25
Yunhui Jo, Young Han, E. Lee, Jaehyeon Seo, Geon Oh, H. Sung, Yongha Gi, Hyun-Woo Kim, Sangmin Park, M. Yoon (2022)
The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3.American journal of cancer research, 12 3
( Ceresoli G , Aerts J , Dziadziuszko R , et al. Tumour treating fields in combination with pemetrexed and cisplatin or carboplatin as first‐line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single‐arm phase 2 trial. Lancet Oncol. 2019;20(12):1702‐1709.31628016)
Ceresoli G , Aerts J , Dziadziuszko R , et al. Tumour treating fields in combination with pemetrexed and cisplatin or carboplatin as first‐line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single‐arm phase 2 trial. Lancet Oncol. 2019;20(12):1702‐1709.31628016Ceresoli G , Aerts J , Dziadziuszko R , et al. Tumour treating fields in combination with pemetrexed and cisplatin or carboplatin as first‐line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single‐arm phase 2 trial. Lancet Oncol. 2019;20(12):1702‐1709.31628016, Ceresoli G , Aerts J , Dziadziuszko R , et al. Tumour treating fields in combination with pemetrexed and cisplatin or carboplatin as first‐line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single‐arm phase 2 trial. Lancet Oncol. 2019;20(12):1702‐1709.31628016
M. Levin, C. Stevenson (2012)
Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering.Annual review of biomedical engineering, 14
Naoyo Nishida, H. Yano, T. Nishida, T. Kamura, M. Kojiro (2006)
Angiogenesis in CancerVascular Health and Risk Management, 2
( Miller R , Song A , Ali A , et al. Scalp‐sparing radiation with concurrent temozolomide and tumor treating fields (SPARE) for patients with newly diagnosed glioblastoma. Front Oncol. 2022;12:896246.35574391)
Miller R , Song A , Ali A , et al. Scalp‐sparing radiation with concurrent temozolomide and tumor treating fields (SPARE) for patients with newly diagnosed glioblastoma. Front Oncol. 2022;12:896246.35574391Miller R , Song A , Ali A , et al. Scalp‐sparing radiation with concurrent temozolomide and tumor treating fields (SPARE) for patients with newly diagnosed glioblastoma. Front Oncol. 2022;12:896246.35574391, Miller R , Song A , Ali A , et al. Scalp‐sparing radiation with concurrent temozolomide and tumor treating fields (SPARE) for patients with newly diagnosed glioblastoma. Front Oncol. 2022;12:896246.35574391
( Chen D , Le S , Hutchinson T , et al. Tumor treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest. 2022;132:0021‐9738.)
Chen D , Le S , Hutchinson T , et al. Tumor treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest. 2022;132:0021‐9738.Chen D , Le S , Hutchinson T , et al. Tumor treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest. 2022;132:0021‐9738., Chen D , Le S , Hutchinson T , et al. Tumor treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest. 2022;132:0021‐9738.
R. Deberardinis, N. Chandel (2020)
We need to talk about the Warburg effectNature Metabolism, 2
( Holdgaard S , Cianfanelli V , Pupo E , et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. 2019;10(1):4176.31519908)
Holdgaard S , Cianfanelli V , Pupo E , et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. 2019;10(1):4176.31519908Holdgaard S , Cianfanelli V , Pupo E , et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. 2019;10(1):4176.31519908, Holdgaard S , Cianfanelli V , Pupo E , et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. 2019;10(1):4176.31519908
( Wen P , Macdonald D , Reardon D , et al. Updated response assessment criteria for high‐grade gliomas: response assessment in neuro‐oncology working group. J Clin Oncol. 2010;28(11):1963‐1972.20231676)
Wen P , Macdonald D , Reardon D , et al. Updated response assessment criteria for high‐grade gliomas: response assessment in neuro‐oncology working group. J Clin Oncol. 2010;28(11):1963‐1972.20231676Wen P , Macdonald D , Reardon D , et al. Updated response assessment criteria for high‐grade gliomas: response assessment in neuro‐oncology working group. J Clin Oncol. 2010;28(11):1963‐1972.20231676, Wen P , Macdonald D , Reardon D , et al. Updated response assessment criteria for high‐grade gliomas: response assessment in neuro‐oncology working group. J Clin Oncol. 2010;28(11):1963‐1972.20231676
Shengchao Xu, C. Luo, Dikang Chen, Lu Tang, Ling Chen, Zhixiong Liu (2022)
Correction: Whole transcriptome and proteome analyses identify potential targets and mechanisms underlying tumor treating fields against glioblastomaCell Death & Disease, 13
( Mohan S , Chawla S , Wang S , et al. Assessment of early response to tumor‐treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience. CNS Oncol. 2016;5(3):137‐144.27076281)
Mohan S , Chawla S , Wang S , et al. Assessment of early response to tumor‐treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience. CNS Oncol. 2016;5(3):137‐144.27076281Mohan S , Chawla S , Wang S , et al. Assessment of early response to tumor‐treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience. CNS Oncol. 2016;5(3):137‐144.27076281, Mohan S , Chawla S , Wang S , et al. Assessment of early response to tumor‐treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience. CNS Oncol. 2016;5(3):137‐144.27076281
G. Ceresoli, J. Aerts, R. Dziadziuszko, R. Ramlau, S. Cedres, J. Meerbeeck, M. Mencoboni, D. Planchard, A. Chella, L. Crinò, M. Krzakowski, J. Rüssel, A. Maconi, L. Gianoncelli, F. Grosso (2019)
Tumour Treating Fields in combination with pemetrexed and cisplatin or carboplatin as first-line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single-arm phase 2 trial.The Lancet. Oncology
( Robinson AJ , Jain A , Sherman HG , et al. Toward hijacking bioelectricity in cancer to develop new bioelectronic medicine. Adv Therapeut. 2021;4(3):2000248.)
Robinson AJ , Jain A , Sherman HG , et al. Toward hijacking bioelectricity in cancer to develop new bioelectronic medicine. Adv Therapeut. 2021;4(3):2000248.Robinson AJ , Jain A , Sherman HG , et al. Toward hijacking bioelectricity in cancer to develop new bioelectronic medicine. Adv Therapeut. 2021;4(3):2000248., Robinson AJ , Jain A , Sherman HG , et al. Toward hijacking bioelectricity in cancer to develop new bioelectronic medicine. Adv Therapeut. 2021;4(3):2000248.
A. Haj, C. Doenitz, C. Hohenberger, A. Brawanski, M. Proescholdt (2018)
P01.144 TTFields for newly diagnosed Glioblastoma: Impact of consultation strategyNeuro-Oncology
( Norden A , Young G , Setayesh K , et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787.18316689)
Norden A , Young G , Setayesh K , et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787.18316689Norden A , Young G , Setayesh K , et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787.18316689, Norden A , Young G , Setayesh K , et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787.18316689
Weixing Zhao, J. Steinfeld, Fengshan Liang, Xiaoyong Chen, D. Maranon, C. Ma, Youngho Kwon, T. Rao, Weibin Wang, chen Sheng, Xuemei Song, Yanhong Deng, J. Jimenez-Sainz, Lucy Lu, R. Jensen, Y. Xiong, G. Kupfer, C. Wiese, E. Greene, P. Sung (2017)
BRCA1–BARD1 promotes RAD51-mediated homologous DNA pairingNature, 550
( Egeblad M , Werb Z . New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174.11990853)
Egeblad M , Werb Z . New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174.11990853Egeblad M , Werb Z . New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174.11990853, Egeblad M , Werb Z . New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174.11990853
L. Lazaridis, E. Bumes, Dorothee Spille, T. Schulz, S. Heider, S. Agkatsev, T. Schmidt, T. Blau, C. Oster, J. Feldheim, W. Stummer, A. Kessler, C. Seidel, O. Grauer, P. Hau, U. Sure, K. Keyvani, U. Herrlinger, C. Kleinschnitz, M. Stuschke, K. Herrmann, C. Deuschl, Stella Breuer, E. Hattingen, B. Scheffler, S. Kebir, M. Glas (2022)
First multicentric real-life experience with the combination of CCNU and temozolomide in newly diagnosed MGMT promoter methylated IDH wildtype glioblastomaNeuro-Oncology Advances, 4
L. Lazaridis, N. Schäfer, Sarah Teuber-Hanselmann, T. Blau, T. Schmidt, C. Oster, J. Weller, T. Tzaridis, D. Pierscianek, K. Keyvani, C. Kleinschnitz, M. Stuschke, B. Scheffler, C. Deuschl, U. Sure, U. Herrlinger, S. Kebir, M. Glas (2019)
Tumour Treating Fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastomaJournal of Cancer Research and Clinical Oncology, 146
Susan Kline-Smith, C. Walczak (2004)
Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics.Molecular cell, 15 3
( Tsujimoto Y , Shimizu S . Another way to die: autophagic programmed cell death. Cell Death Diff. 2005;12(2):1528‐1534.)
Tsujimoto Y , Shimizu S . Another way to die: autophagic programmed cell death. Cell Death Diff. 2005;12(2):1528‐1534.Tsujimoto Y , Shimizu S . Another way to die: autophagic programmed cell death. Cell Death Diff. 2005;12(2):1528‐1534., Tsujimoto Y , Shimizu S . Another way to die: autophagic programmed cell death. Cell Death Diff. 2005;12(2):1528‐1534.
A. Green, Jean Levy, Rajeev Vibhakar, Molly Hemenway, J. Madden, N. Foreman, K. Dorris (2017)
Tumor treating fields in pediatric high-grade gliomaChild's Nervous System, 33
A. Davies, U. Weinberg, Y. Palti (2013)
Tumor treating fields: a new frontier in cancer therapyAnnals of the New York Academy of Sciences, 1291
M. Giladi, M. Munster, R. Schneiderman, T. Voloshin, Y. Porat, R. Blat, Katarzyna Zielinska-Chomej, P. Hååg, Z. Bomzon, E. Kirson, U. Weinberg, K. Viktorsson, R. Lewensohn, Y. Palti (2017)
Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cellsRadiation Oncology, 12
Ryan Miller, A. Song, Ayesha Ali, M. Niazi, V. Bar-ad, N. Martinez, J. Glass, I. Alnahhas, D. Andrews, K. Judy, J. Evans, C. Farrell, M. Werner-Wasik, I. Chervoneva, M. Ly, J. Palmer, Haisong Liu, W. Shi (2022)
Scalp-Sparing Radiation With Concurrent Temozolomide and Tumor Treating Fields (SPARE) for Patients With Newly Diagnosed GlioblastomaFrontiers in Oncology, 12
( Kutuk T , Appel H , Avendano M , et al. Feasibility of tumor treating fields with pemetrexed and platinum‐based chemotherapy for unresectable malignant pleural mesothelioma: single‐center, real‐world data. Cancer. 2022;14(8):2020.)
Kutuk T , Appel H , Avendano M , et al. Feasibility of tumor treating fields with pemetrexed and platinum‐based chemotherapy for unresectable malignant pleural mesothelioma: single‐center, real‐world data. Cancer. 2022;14(8):2020.Kutuk T , Appel H , Avendano M , et al. Feasibility of tumor treating fields with pemetrexed and platinum‐based chemotherapy for unresectable malignant pleural mesothelioma: single‐center, real‐world data. Cancer. 2022;14(8):2020., Kutuk T , Appel H , Avendano M , et al. Feasibility of tumor treating fields with pemetrexed and platinum‐based chemotherapy for unresectable malignant pleural mesothelioma: single‐center, real‐world data. Cancer. 2022;14(8):2020.
M. Tian, Xuexin He, Chentao Jin, Xiao He, Shuang Wu, Rui Zhou, Xiaohui Zhang, Kai Zhang, W. Gu, Jing Wang, Hong Zhang (2021)
Transpathology: molecular imaging-based pathologyEuropean Journal of Nuclear Medicine and Molecular Imaging, 48
Toshiaki Inoue, Y. Nakayama, Yanze Li, Haruka Matsumori, Haruka Takahashi, H. Kojima, H. Wanibuchi, M. Katoh, M. Oshimura (2014)
SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitorsThe FEBS Journal, 281
( Lin Y , Chen B . Case report: tumor‐treating fields prolongs IDH‐mutant anaplastic astrocytoma progression‐free survival and pathological evolution to glioblastoma. Ann Transl Med. 2021;9(24):1804.35071498)
Lin Y , Chen B . Case report: tumor‐treating fields prolongs IDH‐mutant anaplastic astrocytoma progression‐free survival and pathological evolution to glioblastoma. Ann Transl Med. 2021;9(24):1804.35071498Lin Y , Chen B . Case report: tumor‐treating fields prolongs IDH‐mutant anaplastic astrocytoma progression‐free survival and pathological evolution to glioblastoma. Ann Transl Med. 2021;9(24):1804.35071498, Lin Y , Chen B . Case report: tumor‐treating fields prolongs IDH‐mutant anaplastic astrocytoma progression‐free survival and pathological evolution to glioblastoma. Ann Transl Med. 2021;9(24):1804.35071498
( Elzinga G , Wong E . Resolution of cystic enhancement to add‐on tumor treating electric fields for recurrent glioblastoma after incomplete response to bevacizumab. Case Reports Neurol. 2014;6(1):109‐115.)
Elzinga G , Wong E . Resolution of cystic enhancement to add‐on tumor treating electric fields for recurrent glioblastoma after incomplete response to bevacizumab. Case Reports Neurol. 2014;6(1):109‐115.Elzinga G , Wong E . Resolution of cystic enhancement to add‐on tumor treating electric fields for recurrent glioblastoma after incomplete response to bevacizumab. Case Reports Neurol. 2014;6(1):109‐115., Elzinga G , Wong E . Resolution of cystic enhancement to add‐on tumor treating electric fields for recurrent glioblastoma after incomplete response to bevacizumab. Case Reports Neurol. 2014;6(1):109‐115.
G. Ceccon, L. Lazaridis, G. Stoffels, M. Rapp, Manuel Weber, T. Blau, P. Lohmann, S. Kebir, K. Herrmann, G. Fink, K. Langen, M. Glas, N. Galldiks (2018)
Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour-treating fields: initial experienceEuropean Journal of Nuclear Medicine and Molecular Imaging, 45
( Tooley A , Gilden J , Jacobelli J , et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol. 2009;11(1):17‐26.19043408)
Tooley A , Gilden J , Jacobelli J , et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol. 2009;11(1):17‐26.19043408Tooley A , Gilden J , Jacobelli J , et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol. 2009;11(1):17‐26.19043408, Tooley A , Gilden J , Jacobelli J , et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol. 2009;11(1):17‐26.19043408
Y. Tsujimoto, S. Shimizu (2005)
Another way to die: autophagic programmed cell deathCell Death and Differentiation, 12
( Bosnyák E , Barger G , Michelhaugh S , et al. Amino acid PET imaging of the early metabolic response during tumor‐treating fields (TTFields) therapy in recurrent glioblastoma. Clin Nucl Med. 2018;43(3):176‐179.29261637)
Bosnyák E , Barger G , Michelhaugh S , et al. Amino acid PET imaging of the early metabolic response during tumor‐treating fields (TTFields) therapy in recurrent glioblastoma. Clin Nucl Med. 2018;43(3):176‐179.29261637Bosnyák E , Barger G , Michelhaugh S , et al. Amino acid PET imaging of the early metabolic response during tumor‐treating fields (TTFields) therapy in recurrent glioblastoma. Clin Nucl Med. 2018;43(3):176‐179.29261637, Bosnyák E , Barger G , Michelhaugh S , et al. Amino acid PET imaging of the early metabolic response during tumor‐treating fields (TTFields) therapy in recurrent glioblastoma. Clin Nucl Med. 2018;43(3):176‐179.29261637
( Jo Y , Han Y , Lee E , et al. The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3. Am J Cancer Res. 2022;12(3):1423‐1432.35411245)
Jo Y , Han Y , Lee E , et al. The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3. Am J Cancer Res. 2022;12(3):1423‐1432.35411245Jo Y , Han Y , Lee E , et al. The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3. Am J Cancer Res. 2022;12(3):1423‐1432.35411245, Jo Y , Han Y , Lee E , et al. The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3. Am J Cancer Res. 2022;12(3):1423‐1432.35411245
A. Korshoej, S. Lukacova, Y. Lassen-Ramshad, C. Rahbek, K. Severinsen, T. Guldberg, N. Mikic, Mette Jensen, S. Cortnum, G. Oettingen, J. Sørensen (2020)
OptimalTTF-1: Enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastomaNeuro-oncology Advances, 2
E. Kirson, V. Dbalý, F. Tovaryš, J. Vymazal, J. Soustiel, A. Itzhaki, D. Mordechovich, Shirley Steinberg-Shapira, Zoya Gurvich, R. Schneiderman, Yoram Wasserman, M. Salzberg, B. Ryffel, D. Goldsher, E. Dekel, Y. Palti (2007)
Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumorsProceedings of the National Academy of Sciences, 104
( Fisher J , Adamson D . Current FDA‐approved therapies for high‐grade malignant gliomas. Biomedicine. 2021;9(3):324.)
Fisher J , Adamson D . Current FDA‐approved therapies for high‐grade malignant gliomas. Biomedicine. 2021;9(3):324.Fisher J , Adamson D . Current FDA‐approved therapies for high‐grade malignant gliomas. Biomedicine. 2021;9(3):324., Fisher J , Adamson D . Current FDA‐approved therapies for high‐grade malignant gliomas. Biomedicine. 2021;9(3):324.
C. Arvanitis, G. Ferraro, R. Jain (2019)
The blood–brain barrier and blood–tumour barrier in brain tumours and metastasesNature Reviews Cancer, 20
Ute Probst, Irene Fuhrmann, L. Beyer, P. Wiggermann (2018)
Electrochemotherapy as a New Modality in Interventional Oncology: A ReviewTechnology in Cancer Research & Treatment, 17
M. Mehta, V. Gondi, M. Ahluwalia, P. Brown (2019)
Radiosurgery followed by tumour treating fields (TTFields) for brain metastases (1-10) from NSCLC in the phase III METIS trialAnnals of Oncology
Dongjiang Chen, S. Le, Tarun Hutchinson, Anda-Alexandra Calinescu, M. Sebastian, Dan Jin, Tianyi Liu, A. Ghiaseddin, Maryam Rahman, D. Tran (2022)
Tumor Treating Fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastomaThe Journal of Clinical Investigation, 132
( Gera N , Yang A , Holtzman T , Lee S , Wong E , Swanson K . Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PloS One. 2015;10(5):e0125269.26010837)
Gera N , Yang A , Holtzman T , Lee S , Wong E , Swanson K . Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PloS One. 2015;10(5):e0125269.26010837Gera N , Yang A , Holtzman T , Lee S , Wong E , Swanson K . Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PloS One. 2015;10(5):e0125269.26010837, Gera N , Yang A , Holtzman T , Lee S , Wong E , Swanson K . Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PloS One. 2015;10(5):e0125269.26010837
(2020)
Tumor treating fields (TTFields) hinder cancer cell motility through regulation of microtubule and acting dynamics
Q. Ostrom, H. Gittleman, Peter Liao, Chaturia Rouse, Yanwen Chen, J. Dowling, Yingli Wolinsky, C. Kruchko, J. Barnholtz-Sloan (2012)
CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007-2011.Neuro-oncology, 16 Suppl 4
M. Giladi, R. Schneiderman, T. Voloshin, Y. Porat, M. Munster, R. Blat, Shay Sherbo, Z. Bomzon, N. Urman, A. Itzhaki, S. Cahal, A. Shteingauz, A. Chaudhry, E. Kirson, U. Weinberg, Y. Palti (2015)
Mitotic Spindle Disruption by Alternating Electric Fields Leads to Improper Chromosome Segregation and Mitotic Catastrophe in Cancer CellsScientific Reports, 5
J. Baehring, K. Becker (2012)
Faculty Opinions recommendation of NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality.
( Nishida N , Yano H , Nishida T , Kamura T , Kojiro M . Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219.17326328)
Nishida N , Yano H , Nishida T , Kamura T , Kojiro M . Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219.17326328Nishida N , Yano H , Nishida T , Kamura T , Kojiro M . Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219.17326328, Nishida N , Yano H , Nishida T , Kamura T , Kojiro M . Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219.17326328
( Kais Z , Rondinelli B , Holmes A , et al. FANCD2 maintains fork stability in BRCA1/2‐deficient tumors and promotes alternative end‐joining DNA repair. Cell Rep. 2016;15(11):2488‐2499.27264184)
Kais Z , Rondinelli B , Holmes A , et al. FANCD2 maintains fork stability in BRCA1/2‐deficient tumors and promotes alternative end‐joining DNA repair. Cell Rep. 2016;15(11):2488‐2499.27264184Kais Z , Rondinelli B , Holmes A , et al. FANCD2 maintains fork stability in BRCA1/2‐deficient tumors and promotes alternative end‐joining DNA repair. Cell Rep. 2016;15(11):2488‐2499.27264184, Kais Z , Rondinelli B , Holmes A , et al. FANCD2 maintains fork stability in BRCA1/2‐deficient tumors and promotes alternative end‐joining DNA repair. Cell Rep. 2016;15(11):2488‐2499.27264184
( Ostrom QT , Gittleman H , Truitt G , Boscia A , Kruchko C , Barnholtz‐Sloan JS . CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86.30445539)
Ostrom QT , Gittleman H , Truitt G , Boscia A , Kruchko C , Barnholtz‐Sloan JS . CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86.30445539Ostrom QT , Gittleman H , Truitt G , Boscia A , Kruchko C , Barnholtz‐Sloan JS . CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86.30445539, Ostrom QT , Gittleman H , Truitt G , Boscia A , Kruchko C , Barnholtz‐Sloan JS . CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86.30445539
M. Mehta, P. Wen, R. Nishikawa, D. Reardon, K. Peters (2017)
Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients.Critical reviews in oncology/hematology, 111
( Salvador E , Köppl T , Hörmann J , et al. Tumor treating fields (TTFields) induce cell junction alterations in a human 3D In vitro model of the blood‐brain barrier. Pharmaceutics. 2023;15(1):185.36678814)
Salvador E , Köppl T , Hörmann J , et al. Tumor treating fields (TTFields) induce cell junction alterations in a human 3D In vitro model of the blood‐brain barrier. Pharmaceutics. 2023;15(1):185.36678814Salvador E , Köppl T , Hörmann J , et al. Tumor treating fields (TTFields) induce cell junction alterations in a human 3D In vitro model of the blood‐brain barrier. Pharmaceutics. 2023;15(1):185.36678814, Salvador E , Köppl T , Hörmann J , et al. Tumor treating fields (TTFields) induce cell junction alterations in a human 3D In vitro model of the blood‐brain barrier. Pharmaceutics. 2023;15(1):185.36678814
Y. Palti (1962)
Stimulation of muscles and nerves by means of externally applied electrodes.Bulletin of the Research Council of Israel. Section E: Experimental medicine, 10
Nidhi Gera, Aaron Yang, Talia Holtzman, S. Lee, E. Wong, K. Swanson (2015)
Tumor Treating Fields Perturb the Localization of Septins and Cause Aberrant Mitotic ExitPLoS ONE, 10
( Korshoej A , Lukacova S , Lassen‐Ramshad Y , et al. OptimalTTF‐1: enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neuro‐Oncol Adv. 2020;2(1):vdaa121.)
Korshoej A , Lukacova S , Lassen‐Ramshad Y , et al. OptimalTTF‐1: enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neuro‐Oncol Adv. 2020;2(1):vdaa121.Korshoej A , Lukacova S , Lassen‐Ramshad Y , et al. OptimalTTF‐1: enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neuro‐Oncol Adv. 2020;2(1):vdaa121., Korshoej A , Lukacova S , Lassen‐Ramshad Y , et al. OptimalTTF‐1: enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neuro‐Oncol Adv. 2020;2(1):vdaa121.
Yunhui Jo, Eun Kim, S. Sai, Jin Kim, Jae-Min Cho, Hyeongi Kim, J. Baek, Jeong-Yub Kim, Sang-Gu Hwang, M. Yoon (2018)
Functional Biological Activity of Sorafenib as a Tumor-Treating Field Sensitizer for Glioblastoma TherapyInternational Journal of Molecular Sciences, 19
( Achrol A , Rennert R , Anders C , et al. Brain metastases. Nat Rev Dis Prim. 2019;5(1):5.30655533)
Achrol A , Rennert R , Anders C , et al. Brain metastases. Nat Rev Dis Prim. 2019;5(1):5.30655533Achrol A , Rennert R , Anders C , et al. Brain metastases. Nat Rev Dis Prim. 2019;5(1):5.30655533, Achrol A , Rennert R , Anders C , et al. Brain metastases. Nat Rev Dis Prim. 2019;5(1):5.30655533
( Wu H , Yang L , Liu H , et al. Exploring the efficacy of tumor electric field therapy against glioblastoma: an in vivo and in vitro study. CNS Neurosci Ther. 2021;27(12):1587‐1604.34710276)
Wu H , Yang L , Liu H , et al. Exploring the efficacy of tumor electric field therapy against glioblastoma: an in vivo and in vitro study. CNS Neurosci Ther. 2021;27(12):1587‐1604.34710276Wu H , Yang L , Liu H , et al. Exploring the efficacy of tumor electric field therapy against glioblastoma: an in vivo and in vitro study. CNS Neurosci Ther. 2021;27(12):1587‐1604.34710276, Wu H , Yang L , Liu H , et al. Exploring the efficacy of tumor electric field therapy against glioblastoma: an in vivo and in vitro study. CNS Neurosci Ther. 2021;27(12):1587‐1604.34710276
M. Lacouture, M. Davis, G. Elzinga, N. Butowski, D. Tran, J. Villano, L. DiMeglio, A. Davies, E. Wong (2014)
Characterization and management of dermatologic adverse events with the NovoTTF-100A System, a novel anti-mitotic electric field device for the treatment of recurrent glioblastoma.Seminars in oncology, 41 Suppl 4
稲垣 隆介 (2008)
The International Society for Pediatric Neurosurgery に参加して, 33
R. Stupp, Eric Wong, A. Kanner, D. Steinberg, H. Engelhard, V. Heidecke, E. Kirson, S. Taillibert, F. Liebermann, V. Dbalý, Z. Ram, J. Villano, N. Rainov, U. Weinberg, David Schiff, L. Kunschner, J. Raizer, J. Honnorat, A. Sloan, M. Malkin, J. Landolfi, F. Payer, M. Mehdorn, Robert Weil, S. Pannullo, M. Westphal, M. Smrcka, L. Chin, H. Kostron, S. Hofer, Jeffrey Bruce, R. Cosgrove, Nina Paleologous, Y. Palti, P. Gutin (2012)
NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality.European journal of cancer, 48 14
J. Branter, Maria Estevez-Cebrero, Mohammed Diksin, Michael Griffin, M. Castellanos-Uribe, S. May, R. Rahman, R. Grundy, S. Basu, S. Smith (2022)
Genome-Wide Expression and Anti-Proliferative Effects of Electric Field Therapy on Pediatric and Adult Brain TumorsInternational Journal of Molecular Sciences, 23
Q. Ostrom, N. Patil, G. Cioffi, K. Waite, C. Kruchko, J. Barnholtz-Sloan (2020)
CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017.Neuro-oncology, 22 Supplement_1
( Park J , Song K , Jung S , et al. Tumor‐treating fields induce RAW264.7 macrophage activation via NK‐κB/MAPK signaling pathways. Technol Cancer Res Treat. 2019;18:1533033819868225.)
Park J , Song K , Jung S , et al. Tumor‐treating fields induce RAW264.7 macrophage activation via NK‐κB/MAPK signaling pathways. Technol Cancer Res Treat. 2019;18:1533033819868225.Park J , Song K , Jung S , et al. Tumor‐treating fields induce RAW264.7 macrophage activation via NK‐κB/MAPK signaling pathways. Technol Cancer Res Treat. 2019;18:1533033819868225., Park J , Song K , Jung S , et al. Tumor‐treating fields induce RAW264.7 macrophage activation via NK‐κB/MAPK signaling pathways. Technol Cancer Res Treat. 2019;18:1533033819868225.
( Martins I , Wang Y , Michaud M , et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79‐91.23852373)
Martins I , Wang Y , Michaud M , et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79‐91.23852373Martins I , Wang Y , Michaud M , et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79‐91.23852373, Martins I , Wang Y , Michaud M , et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79‐91.23852373
( Lazaridis L , Schäfer N , Teuber‐Hanselmann S , et al. Tumour treating fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastoma. J Cancer Res Clin Oncol. 2020;146(3):787‐792.31828428)
Lazaridis L , Schäfer N , Teuber‐Hanselmann S , et al. Tumour treating fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastoma. J Cancer Res Clin Oncol. 2020;146(3):787‐792.31828428Lazaridis L , Schäfer N , Teuber‐Hanselmann S , et al. Tumour treating fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastoma. J Cancer Res Clin Oncol. 2020;146(3):787‐792.31828428, Lazaridis L , Schäfer N , Teuber‐Hanselmann S , et al. Tumour treating fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastoma. J Cancer Res Clin Oncol. 2020;146(3):787‐792.31828428
T. Kutuk, H. Appel, M. Avendano, F. Albrecht, P. Kaywin, Suyen Ramos, Melanie Suarez-Murias, M. Mehta, R. Kotecha (2022)
Feasibility of Tumor Treating Fields with Pemetrexed and Platinum-Based Chemotherapy for Unresectable Malignant Pleural Mesothelioma: Single-Center, Real-World DataCancers, 14
( Ceccon G , Lazaridis L , Stoffels G , et al. Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour‐treating fields: initial experience. Eur J Nucl Med Mol Imaging. 2018;45(9):1626‐1635.29564490)
Ceccon G , Lazaridis L , Stoffels G , et al. Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour‐treating fields: initial experience. Eur J Nucl Med Mol Imaging. 2018;45(9):1626‐1635.29564490Ceccon G , Lazaridis L , Stoffels G , et al. Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour‐treating fields: initial experience. Eur J Nucl Med Mol Imaging. 2018;45(9):1626‐1635.29564490, Ceccon G , Lazaridis L , Stoffels G , et al. Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour‐treating fields: initial experience. Eur J Nucl Med Mol Imaging. 2018;45(9):1626‐1635.29564490
P. Wen, D. MacDonald, D. Reardon, T. Cloughesy, A. Sorensen, E. Galanis, J. Degroot, W. Wick, M. Gilbert, A. Lassman, C. Tsien, T. Mikkelsen, Eric Wong, M. Chamberlain, R. Stupp, K. Lamborn, M. Vogelbaum, M. Bent, Susan Chang (2010)
Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group.Journal of clinical oncology : official journal of the American Society of Clinical Oncology, 28 11
Hao Wu, Lin Yang, Han-jie Liu, Dan Zhou, Dikang Chen, Xiao-que Zheng, Huijun Yang, Chong Li, Jiusheng Chang, A. Wu, Zhifei Wang, N. Ren, S. Lv, Yuyang Liu, Muyuan Jia, Jian Lu, Hongyu Liu, Guochen Sun, Zhixiong Liu, Jialin Liu, Ling Chen (2021)
Exploring the efficacy of tumor electric field therapy against glioblastoma: An in vivo and in vitro studyCNS Neuroscience & Therapeutics, 27
(2008)
Amoeboid T lymphocytes Require the Septin Cytoskeleton for Cortical Integrity and Persistent MotilityNature cell biology, 11
( Wong E , Lok E , Gautam S , Swanson K . Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113(2):232‐241.26125449)
Wong E , Lok E , Gautam S , Swanson K . Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113(2):232‐241.26125449Wong E , Lok E , Gautam S , Swanson K . Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113(2):232‐241.26125449, Wong E , Lok E , Gautam S , Swanson K . Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113(2):232‐241.26125449
( Davies A , Weinberg U , Palti Y . Tumor treating fields: a new frontier in cancer therapy. Ann N Y Acad Sci. 2013;1291:86‐95.23659608)
Davies A , Weinberg U , Palti Y . Tumor treating fields: a new frontier in cancer therapy. Ann N Y Acad Sci. 2013;1291:86‐95.23659608Davies A , Weinberg U , Palti Y . Tumor treating fields: a new frontier in cancer therapy. Ann N Y Acad Sci. 2013;1291:86‐95.23659608, Davies A , Weinberg U , Palti Y . Tumor treating fields: a new frontier in cancer therapy. Ann N Y Acad Sci. 2013;1291:86‐95.23659608
R. Kalluri, R. Weinberg (2009)
The basics of epithelial-mesenchymal transition.The Journal of clinical investigation, 119 6
Eun Kim, Yeon Kim, Hyo-Sook Song, Youn Jeong, J. Lee, J. Sung, S. Yoo, M. Yoon (2016)
Biological effect of an alternating electric field on cell proliferation and synergistic antimitotic effect in combination with ionizing radiationOncotarget, 7
( Salzberg M , Kirson E , Palti Y , Rochlitz C . A pilot study with very low‐intensity, intermediate‐frequency electric fields in patients with locally advanced and/or metastatic solid tumors. Onkologie. 2008;31(7):362‐365.18596382)
Salzberg M , Kirson E , Palti Y , Rochlitz C . A pilot study with very low‐intensity, intermediate‐frequency electric fields in patients with locally advanced and/or metastatic solid tumors. Onkologie. 2008;31(7):362‐365.18596382Salzberg M , Kirson E , Palti Y , Rochlitz C . A pilot study with very low‐intensity, intermediate‐frequency electric fields in patients with locally advanced and/or metastatic solid tumors. Onkologie. 2008;31(7):362‐365.18596382, Salzberg M , Kirson E , Palti Y , Rochlitz C . A pilot study with very low‐intensity, intermediate‐frequency electric fields in patients with locally advanced and/or metastatic solid tumors. Onkologie. 2008;31(7):362‐365.18596382
INTRODUCTIONMalignant brain tumors are lethal diseases of the central nervous system and pose a serious threat to human health because of the specificity of tumor sites. Among them, glioblastoma multiforme (GBM) is the most common and deadliest primary neoplasm with an incidence of 5/100000 inhabitants/year and a recurrence rate approaching 100%.1,2 The current standard of care (SOC) for GBM is multidisciplinary treatment approaches, such as the Stupp protocol, involving extensive surgery, chemotherapy and radiotherapy, the prognosis remains discouraging, showing a median survival of 15 months and a 5‐year survival rate of less than 5%.3 Temozolomide (TMZ) is an indispensable clinical first‐line chemotherapeutic agent for the treatment of GBM but is limited by drug resistance.4 The conventional approaches provide limited effectiveness for malignant brain tumors due to late diagnosis, unspecific location, and tumor heterogeneity, leading to unbearable adverse effects.5,6 Targeted therapy and immunotherapy (e.g., gefitinib and trastuzumab) have shown modest efficacy, because of the poor permeability of the blood–brain‐barrier (BBB).7–9 Therefore, it's urgent to develop a more effective and safer therapy.TTFields are a novel tumor therapy that inhibits the proliferation and growth of the tumor by delivering alternating electrical field (EF) at low intensity (1–3 V/cm) and intermediate frequency (100–300 kHz).10 TTFields therapy has achieved satisfactory outcomes in several clinical trials. As a physical local therapy, TTFields do not cause serious systemic side effects and are free of therapeutic resistance.11,12 Up to now, TTFields have already been approved by the US Food and Drug Administration (FDA) for the treatment of patients with newly diagnosed glioblastoma (ndGBM), recurrent glioblastoma (rGBM), and malignant pleural mesothelioma,11–13 and recommended by many domestic and international expert consensus and guidelines.Afterward, breakthroughs have been made in the in vitro or animal studies on the biological mechanisms of TTFields, and the potential of TTField‐based combination therapies continues to be explored. Clinicians have carried out some significant trials of TTFields, including combined treatments or triple treatment, making TTFields a promising candidate in combination with other therapies. In this article, we will review the anti‐tumor mechanism of TTFields and discuss the emerging framework of TTFields‐based combination therapy for malignant brain tumors.ANTI‐TUMOR MECHANISMS OF TTFIELDSTissues and cells possess endogenous EF that influence biological activities and cellular events. Bioelectrical signaling regulates many essential processes to cellular homeostasis, and the biological circuitry of cancer cells is modified. Such as the processes of tumor metastasis, which can be regulated by cellular ion channels.14 When potassium ion channels are overexpressed in tumor cells, more negative charges will be carried inside the cells, and the imbalance of voltage will lead to an increase in tumor growth and metastasis. Manipulating the voltage characteristics of breast cancer cells can significantly reduce the number of metastatic sites in the lungs of mice by about 50%.15 Exogenous EF has been long exploited for interference and/or stimulation of certain natural biological processes, such as depolarization of nerves, contraction of muscles, embryonic development and heat production of tissues.16 Neither heat is generated nor action potentials are triggered, so the application of mid‐frequency alternating EF is neglected.17,18 Emerging studies have reported that TTFields can alter the bioelectrical state of macromolecules and organelles involved in cancer biology, thus showcasing the therapeutic potential against tumors. Moreover, the whole proteomic and transcriptomic analyzes proved the massive alteration of differentially expressed proteins, mRNAs, miRNAs, lncRNAs, and circRNAs by TTFields in GBM cells, which are related to cell mitosis‐related events, varied cellular biological processes, and multiple organelle structures and activities.19TTFields interfere with cancer cell mitosisThe inhibition of cell mitosis is the most commonly reported mechanism of TTFields and TTFields target cancer cells through unusual electrical polarity and rapid proliferative properties. Considering that all charged particles and dipoles in the cell will respond to EF/currents and will oscillate as EF forces alternate in opposite directions,20 and dividing cells contain highly polar, spatially oriented microtubules and septins, TTFields are capable of interfering with cell mitosis and leading to the arrest of proliferation.10 Specifically, during normal metaphase, tubulins are precisely choreographed and arranged to form microtubule spindles that extend into the genetic material lining the center of the cell and bind to chromosomes.21,22 When exposed to TTFields, tubulin is forced to align along the direction of EF, resulting in the interference of tubulin polymerization and obstruction of microtubule spindle formation (Figure 1A).101FIGUREDiagram of the anti‐cancer mechanism of TTFields. (A) During metaphase mitosis, tubulin will align in the direction of the EF, resulting in interference with its polymerization and thus impaired chromosome segregation. (B) During cell division, the cell morphology under TTFields (hourglass‐like shape) produces an inhomogeneous intracellular EF, and all polar macromolecules and organelles will be pushed toward the CCF. In addition to interfering with mitosis, TTFields have multiple biological mechanisms, including (C) inhibiting DSB repair, enhancing DNA replication pressure, (D) inhibiting tumor cell migration and metastasis, (E) promoting autophagy, and (F) inflammatory responses to kill tumor cells, (G) increasing cell membrane and (H) BBB permeability to facilitate the uptake of agents.Another important basis for the effect of TTFields in dividing cells is the directional, hourglass‐shaped cell morphology during the cytokinesis phase.10 At the late stage of cell mitosis, the mitotic septin complex composes of septin 2, 6, and 7, which will be repositioned in alternating EF, leading to aberrant localization of the cytokinetic cleavage furrow (CCF), which results in improper cell division.23 Septins cross‐link to F‐actin bundles within the submembrane actin cytoskeleton and must be sufficiently stiff to withstand the hydrostatic pressure generated within the cytoplasm by the invasion of CCF.24,25 In cytokinesis, the applied EF intensity in the dividing cell shows an hourglass‐like non‐uniform field distribution, and all polar macromolecules and organelles will be pushed toward the CCF where the EF intensity is highest (Figure 1B).26–28 This so‐called “mesoelectrophoresis” prevents cells from dividing properly. Catastrophic mitotic errors that occur in late mitosis are impossibly rectified, and the cell membrane will rupture and blister.29 Then cancer cells will undergo cell death or apoptosis. A systematic comparison of changes in cell number and clonogenic survival of 10 solid tumor‐derived cell lines after 72 h of TTFields exposure yielded that all cell lines exhibited a decrease in cell number and clonogenic survival.27 However, the mechanism of apoptosis induced by TTfields has not been elucidated completely. Some studies revealed that TTFields‐induced cell death occurs in a caspase 3‐dependent manner,30 while some argued that TTFields‐induced apoptosis is not dependent on caspase 3.31TTFields disrupt genomic integrityDuring interphase, TTFields possess the function of disrupting the integrity of the genome, leading to efficient cytocidal actions against tumor cells. The phosphorylation level of γ‐H2AX, an indicator of DNA damage, was higher when combining TTFields with radiation treatment (RT) than RT alone.32 RT‐induced cytotoxicity depends on the extent of DNA double‐strand breaks (DSBs) repair, most of which are repaired within 24 h after RT. In TTFields‐treated cells after RT, more than 40% of DSB failed to be repaired, indicating the efficacy of TTFields to enhance RT‐damaged DNA may be through blocking the homologous recombination repair pathway.33 The expression of BRCA1 pathway genes was found significantly down‐regulated during TTFields treatment.34 The BRCA1 pathway not only plays a vital role in homologous recombinant DNA repair,35 but maintains the stability of DNA replication forks in association with the Fanconi anemia proteins and promotes alternative end‐joining DNA repair.36 Therefore, DNA replication stress is increased by TTFields, including the reduction of replication fork speed and the increase in R‐loop formation, resulting in the disruption of DNA integrity.37 In turn, TTFields not only slow DNA damage repair kinetics but also induce replication stress in cancer cells, resulting in cell death (Figure 1C).TTFields inhibit cell migration and invasionTTFields have been reported with the capacity to inhibit the metastatic spread of solid tumors.38 Tumor metastasis is a multi‐step process, including tumor cell invasion of basement membranes and movement to surrounding tissues, intravasation into blood vessels, and spreading to other organ sites. It's well known that microtubules in TTFields‐treated cells tend to align with the EF. Alterations of microtubules lead to the mediation of the GEF‐H1/RhoA/ROCK signaling pathway and the consequent formation of focal adhesions and induction of peripheral actin bundling, thereby hindering the motility of cancer cells (Figure 1D).23 TTField also exert the suppression of ciliogenesis in GBM cell lines, which is related to the development of tumor and resistance to therapy.39Epithelial‐mesenchymal transition (EMT) programs promote the acquisition of aggressive properties by enhancing the motility of cancer cells, damaging the intercellular junctions, and remodeling the extracellular matrix (ECM).40 EMT‐related biomarkers in GBM cells were found significantly affected by TTFields involving a series of potential mechanisms. Remarkably, mesenchymal markers (e.g., vimentin, smooth muscle actin) were down‐regulated, while epithelial markers (e.g., the adherens junction protein E‐cadherin) were up‐regulated, and its loss serves as the core role in the loss of epithelial differentiation.41 ECM is the first tissue barrier to prevent tumor invasion peripherally, which can be degraded by matrix metalloproteinases (MMPs). TTFields can inhibit the degradation of ECM by suppressing the expression of nuclear factor kappa‐B (NF‐κB), a transcription factor that regulates the expression of MMPs.41,42 Besides, the migration and invasion of cancers require an adequate supply of oxygen and nutrients, hence neovascularization is a decisive factor in cancer progression.43 The levels of hypoxia‐inducible factor 1α (HIF1α) and vascular endothelial growth factor (VEGF) were decreased in TTFields‐treated cells, leading to reduced angiogenesis.41 Furthermore, a significant time‐dependent inhibition in PI3K/AKT and MAPK signaling was observed in the TTFields‐treated cells, resulting in a reduction in cell migration and invasion by decreasing EMT‐ and ECM‐ related marker expression and reducing angiogenesis (Figure 1D).41TTFields intervene autophagy processAbnormal mitotic events can invite autophagy to occur (Figure 1E).44 When exposed to TTFields, the expression of autophagosome marker LC‐II/LC‐I was increased and the cells exhibited typical signs of autophagy.31 Akt2/mTOR/p70S6K axis (a negative regulator of autophagy) is a vital regulator of autophagy by TTFields. Moreover, many miRNAs can be induced by TTFields, especially miR‐29b which directly targets Akt2 to trigger autophagy.45 In most cases, autophagy can maintain the organization and stability of the centrosome to protect cells,46 while in the presence of the autophagy inhibitor 3‐methyladenine, the number of dead GBM cells treated with TTFields was diminished, indicating that autophagy‐mediated TTFields‐induced cell death.31 Such called “autophagic programmed death” may be an alternative to programmed cell death.47However, inconsistent findings have been reported. Some studies proposed that autophagic flux might be interrupted by TTFields.19 Autophagy may be a protective mechanism of cells against TTFields in some cases. Phosphorylated AMP‐dependent kinase (AMPK) can inhibit mTORC1, thereby suppressing its ability to negatively regulate autophagy. Depletion of AMPK inhibits the up‐regulation of autophagy in response to TTFields and sensitizes GBM cells to treatment.48 It is unknown whether autophagy exerts a protective or killing effect on TTFields‐treated cancer cells and may be relevant to the genetic traits of cancer cells, which remains to be further investigated.TTFields induce an anti‐tumor immune responseSystemic anti‐tumor immune responses can be activated in TTFields‐treated tumor‐bearing animals (Figure 1F).38 High doses of dexamethasone (a steroid used to relieve inflammation) could interfere with the therapeutic effect of TTFields on rGBM.49 In TTFields‐treated cells, damage‐related molecular patterns including high‐mobility group box 1 (HMGB1) and ATP were released, and calreticulin was exposed on the cell surface leading to increased infiltration of antigen‐presenting cells into the tumor site. Endoplasmic reticulum stress induced by TTFields may be the trigger that drives exposure of calreticulin to the cell surface.50 The dying cells release ATP, which serves as a “find me” signal for apoptotic cells to increase the recruitment of lymphocytes to induce immunogenic cell death.51 Furthermore, cell death caused by TTFields can promote the maturation and phagocytosis of dendritic cells.50Emerging evidence suggested that TTFields fostered the activation of RAW 264.7 macrophages and its output of NO and ROS. When co‐cultured with 4TI cells (a breast cancer cell line) under TTFields, macrophages secreted elevated levels of pro‐inflammatory cytokines, like IL‐1β, TNF‐α, and IL‐6, and the viability of 4TI cells was diminished. Besides, the phosphorylation levels of IκB‐α, NF‐κB p65 subunit, and p38 MAPK were observed higher in TTFields‐treated RAW 264.7 cells than in control, indicating that TTFields induced the p38 MAPK/NF‐κB pathway of macrophages to exert inflammatory effects.52Recently, TTFields were reported to cause local disruption of the nuclear envelope of cancer cells during interphase, resulting in the release of large cytoplasmic clusters of naked micronuclei, which recruited and activated two major DNA sensors (cGAS and AIM2). Subsequently, the activated cGAS/STING inflammasome tended to up‐regulate pro‐inflammatory cytokines, type 1 interferons (T1IFNs) and T1IFN‐responsive genes, thereby activating the peripheral immune system and creating a potential intrinsic immune platform for cancers53 (Figure 1C).TTFields increase cell membrane permeabilityWhen applying high‐pulsed EF to the cells, depending on the field intensity, irreversible electroporation, induction of necrotic cell death, or reversible electroporation may occur.54 In TTFields‐treated glioblastoma cells, the number and size of membrane pores were reversibly increased, allowing greater permeability to substances as large as 20 kDa (e.g., 5‐aminolevulinic acid) but not exceeding 50 kDa (Figure 1G).55 Interestingly, the effect was tumor‐specific and did not appear on normal cells. EF‐induced transient increase in plasma membrane permeability, with contributions from structural rearrangement of lipids and protein changes, causing fatigue of the membrane structure.54 Since the membrane composition of cancer cells is altered and more deformable than normal cells that may help in explaining the inconsistent response to alternating EF.54 The cell membrane permeability of tumor cells increases in response to TTFields, allowing transmembrane transport of chemical agents that explain the increased efficacy of other drugs to some extent. The cell‐membrane permeabilization also contributes to the release of innate tumor antigens and activates the immune system.56Ion channels in the cell membrane can be affected by alternating EF, and the L‐type Ca2+ channels CACNA1C (Cav1.2) were identified as TTFields target recently,57 thereby influencing cell cycle progression, cell migration, and clonogenic survival of GBM cells.58 Cell membrane potential is a key factor controlling the switching on and off of ion channels in cell membranes, and has been identified as a target for TTFields. Employing the Schwan equation (used to calculate transmembrane potential); it was found that in tumor cells, TTFields‐induced changes in cell membrane potential can be much higher than 10% of the resting cell membrane potential, thus affecting intracellular ion homeostasis.59TTFields were also reported to disrupt tight junction proteins (e.g., claudin 5 and ZO‐1) of the BBB to increase the permeability of the brain (Figure 1H).60 Till now, the presence of BBB has posed limitations for the treatment of brain malignancies and the cerebral application of some agents. Recently, TTFields have been proven to temporarily increase the permeability of the BBB in a human in vitro model.61 TTField‐induced opening of BBB is similarly reversible and expands the range of intracranial drug applications.FACTORS AFFECTING THE EFFICACY OF TTFIELDSThe efficacy of TTFields depends on specific parameters. For different types and sites of tumors, different modalities are set to obtain optimal efficacy. The optimal intensity of EF is different for different cancer cells. Mouse melanoma cells and rat glioma cells completely stop proliferating at 1.35 and 2.25 V/cm respectively,10 while human non‐small cell lung cancer and breast cancer cells require higher.26 The frequency range for anti‐tumor effects in most cancers is 100 to 200 kHz62,63 (e.g., 200 kHz for human GBM clinically, 120 kHz for melanoma cells10). In vitro studies have shown that different GBM cell lines have different optimal electrical frequencies, 200 kHz for KNS42 and GIN‐31, 400 kHz for SF18, and 100 kHz for U87.64,65 The optimal frequency of alternating EF is inversely proportional to the size of the cancer cell.10 Additionally, the inhibitory effect of tumor proliferation by TTFields is time‐dependent,27,66 and the effect of EF is strongest when the direction of EF is parallel to the axis of cell division. Given that the polarity of tumor cells during the division cycle is irregular, applying EF in different directions is an effective way to increase treatment efficacy for patients.10 Sequential application of multiple EF directions every 0.25 ~ 1.00 s can make the splitting axis of more cells parallel to the direction of EF.26CLINICAL STUDIES OF TTFIELDS IN MALIGNANT BRAIN TUMORSWith an in‐depth understanding of the mechanism of action of TTFields, translational studies accompanying clinical trials are crucial for TTFields and are being vigorously pursued. The results of published clinical scientific research on TTFields therapy in brain malignancies have been summarized in Table 1. The ongoing clinical trials to further evaluate the safety and efficacy of TTFields‐based combination therapy have been listed in Table 2.1TABLEThe summary of finished clinical studies of TTFields in brain tumors.StudyPhaseCancer typeNumber of patientsThe device, frequency, intensity, and exposure time of TTFieldsCombining therapyOutcomesRefs.EF‐02Pilot studyrGBM1NovoTTF‐100A device;Frequency: 100–200 kHz; Field intensity of 0.7 V/cm root mean square (RMS).—No response67EF‐07Pilot clinical trialrGBM10Not available.—Median time to disease progression: 26.1 weeks; Mean OS: 62.2 weeks26EF‐11Randomized phase III clinical trialrGBMTTFields arm: 120; TMZ arm: 117NovoTTF‐100A; Frequence: 200 kHz; Field intensity:>0.7 V/cm at the center of the brain.—Mean OS: 6.6 vs. 6.0 months;Mean PFS: 2.2 vs. 2.1 months.11—Retrospective StudyrGBMTMZ/BEV/IRI/TTFields arm: 18;BEV/TTFields arm: 30NovoTTF‐100A; Exposure time: >18 h/day.TMZBEV,IrinotecanMean OS: 18.9 months;Mean PFS: 10.7 months.68OptimalTTF‐1Pilot studyrGBM15Optune; Frequence: 200 kHz; a peak‐to‐peak current of 1.8–2 A.Skull remodeling surgeryMean OS: 15.5 months;Mean PFS: 4.6 months.69EF‐14Randomized phase III clinical trialndGBMTMZ/TTFields arm: 466;TMZ arm: 229NovoTTF‐100A; Frequency: 200 kHz; Exposure time: ≥18 h/day.TMZMean OS: 19.6 vs. 16.6 months;Mean PFS: 7.1 vs. 4.0 months.12—Clinical trialnewly diagnosed MGMT promoter methylated glioblastomaCCNU/TMZ/TTFields≥8 weeks arm: 22CCNU/TMZ/TTFields<8 weeks arm: 48Not available.CCNU, TMZMean PFS: 21.5 vs. 11.2 months;Mean OS: not reached.70—Retrospective StudyndGBM16Optune; ≥18 h/dayTMZ,LomustineMean PFS: 20 months.71—Pilot studyndGBM30NovoTTF‐200A; Frequency: 200 kHz; Exposure time: ≥18 h/day.Concurrent radiotherapy,TMZ.Mean PFS: 9.3 months.72—Pilot studyndGBM10NovoTTF‐200A; Frequency: 200 kHz; Exposure time: ≥18 h/day.Radiotherapy,TMZMean PFS: 8.9 months.73Abbreviations: BEV, Bevacizumab; GBM, glioblastoma; ndGBM, newly‐diagnosed glioblastoma; OS, overall survival; PFS, progression‐free survival; rGBM, recurrent glioblastoma; TMZ, Temozolomide.2TABLEThe summary of ongoing clinical trials of TTFields in brain tumors.NCT numberCondition or diseaseThe device, frequency, intensity, and exposure time of TTFieldsCombining therapyStatusNCT02831959Non‐small cell lung cancer brain metastasisNovoTTF‐200A; 150 kHz; >18 h/dayStereotactic radio surgeryRecruitingNCT05341349Melanoma brain metastasesNovoTTF‐100 M; ≥8 h/dayIpilimumab, Nivolumab, Pembrolizumab, Stereotactic, RadiosurgeryNot yet recruitingNCT04967027Brain metastasis with drug/radiation resistantASCLU‐300 TTF;≥18 h/day—RecruitingNCT04221503GBMOptune; ≥18 h/dayNiraparib, Planned surgical resectionRecruitingNCT04689087rGBMNot mentionedSecond‐line chemotherapy treatmentRecruitingNCT04492163rGBMOptune; 200 kHz—RecruitingNCT04223999First recurrent GBM≥18 h/daySkullremodeling surgeryRecruitingNCT04397679ndGBM≥18 h/dayPartial Brain radiotherapy, Temozolomide, ChloroquineRecruitingNCT03405792ndGBMOptune; 24 monthsTemozolomide, PembrolizumabActive, not recruitingNCT03223103ndGBMOptunePoly‐ICLC, PeptidesActive, not recruitingNCT04671459GBM and rGBMOptuneStereotactic radiosurgeryRecruitingNCT05030298Malignant GliomaNot availableRT, TMZ, radiosurgeryNot yet recruitingNCT04902586Glioma and GBMLow‐intensity; 200 kHz; ≥18 h/dayRadiotherapyNot yet recruitingNCT04396860Gliosarcoma and MGMT‐unmethylated GBMNovoTTF‐100AIpilimumab, NivolumabRecruitingNCT03033992Pediatric malignant glioma ependymomaNovoTTF‐200A; ≥18 h/day—RecruitingNCT05086497GBMOptune array layout mapping created from conventional/advanced MR imaging sequences—Not yet recruitingNote: All trials can be searched on the ClinicalTrials.gov beta website (https://clinicaltrials.gov/).Recurrent glioblastomaDefined as a grade IV glioma, GBM has a high recurrence rate, and there is no consensus for the optimal treatment of rGBM.74,75 Bevacizumab (BEV), lomustine, carmustine and carmustine wafers are FDA approved for rGBM. GBM diffusely infiltrates the brain and rarely metastasizes extracranially, thus is amenable to TTFields therapy with complete coverage of the brain volume.76 The pilot trial of TTFields therapy was on 10 patients with rGBM, and the median time to disease progression was 26.1 weeks and the mean overall survival (OS) was 62.2 weeks, higher than the reported medians of historical control patients.26 In the randomized phase III clinical trial (EF‐11, NCT00379470), 237 patients with rGBM were enrolled into two arms: TTFields versus best physician's choice chemotherapy (including BEV, irinotecan, nitrosurea, carboplatin and TMZ). The results showed that OS and progression‐free survival (PFS) of patients in two arms were similar (6.6 vs. 6.0 months, 2.2 vs. 2.1 months, respectively; p > .05).11 Although no improvement was observed in the EF‐11 trial, the better quality of life (QoL) and lighter toxicity favored the TTFields when compared with chemotherapy as an option in the maintenance phase. Of note, most of the recruited patients were in the advanced stages of the disease (failure of more than 2 chemotherapy agents), and there was heterogeneity in the patient population. In the OptimalTTF‐1 trial (NCT02893137), the combination of cranial remodeling surgery with TTFields and BPC in patients with rGBM yielded an OS of 15.5 months and PFS of 4.6 months,69 much higher than that of TTFields arm in EF‐11, and was well tolerated by patients.11 The removal of a standard craniotomy bone flap increased the EF intensity at the tumor site and was more effective with multiple smaller burr holes than with a single craniectomy.69 Since the new multimodal approach has shown a preliminary survival benefit, a large‐scale randomized, controlled trial, phase 2 trial (OptimalTTF‐2, NCT0422399) was initiated 2 years ago to valid the novel combination therapy.77New diagnosed glioblastomaThe SOC for ndGBM is maximal surgical resection, 6 weeks of postoperative RT and TMZ, and 6 months of TMZ maintenance therapy.78 In a phase III trial of ndGBM (EF‐14, NCT00916409), 695 patients after completion of chemoradiotherapy were enrolled into two arms randomly. Compared with standard TMZ maintenance therapy alone, adding TTFields to standard TMZ maintenance therapy dramatically improved OS (19.6 vs. 16.6 months, respectively; p < .001) and PFS (7.1 vs. 4.0 months, respectively; p < .001) of patients.12 Excitingly enough, the DNA repair protein O6‐methyl‐guanine DNA methyltransferase (MGMT), which has previously been reported to affect the efficacy of TMZ, did not affect TTFields.31 EF‐14 is the landmark trial of TTFields therapy, however, the lack of a sham device control group in this clinical trial may have led to a placebo effect. Moreover, the patient selection in EF‐14 that enrolled patients after standard chemoradiotherapy rather than at diagnosis may lead to some bias, and the timing of TTFields therapy still needs to be determined again. The NOA‐09/CeTeG trial published recently has verified the feasibility of a combination of lomustine and TMZ.71 Therefore, the combination of TTFields, lomustine and TMZ during the maintenance phase was tested, and the observed PFS was 20 months in 16 patients with MGMT promoter methylated ndGBM that might suggest the potential benefit of the triple maintenance therapy.71 A new publishment reported that the combination of TTFields and TMZ plus CCNU can provide additional survival benefits for newly diagnosed MGMT promoter methylated glioblastoma patients. Median PFS of 14.4 months and median OS of 33.8 months were observed in 70 patients.70TTFields are a promising candidate radiosensitizer which induced an abnormal increase in mitotic catastrophe and DNA damage of cells.32 A prospective, single‐arm study that recruited 10 patients with ndGBM was conducted to assess the feasibility and safety of combined RT and TTFields therapy with maintenance TMZ and TTFields. The mean PFS of this trial was 8.9 months and limited toxicity was reported.73 When TTFields were given concurrently with RT which was delivered through the TTFields arrays and with maintenance TMZ, the PFS of 9.3 months was illustrated.72 It follows that the feasibility of TTFields both as maintenance therapy and alongside chemoradiotherapy is endorsed by prior clinical practice, and a more large‐scale clinical trial, EF‐32(NCT04471844), is underway.Other gliomasThe experience with the application of TTFields to other gliomas is limited. A published clinical case for the first time reported that TTFields‐based combination therapy delayed pathological up‐gradation from anaplastic astrocytoma (a WHO grade III glioma) to GBM and prolonged PFS to nearly 10 months.79 This case indicated the potential benefit of TTFields‐based therapy in anaplastic astrocytoma and additional experiments of TTFields in a larger cohort of patients with anaplastic astrocytoma needed to be evaluated.Ganglioglioma is classified as grade I, and although grows slowly, some patients will experience recurrence or malignant progression. Transformation of ganglioglioma to high‐grade glioma is rare but usually with a poor prognosis.80 A patient with a BRAF V600E mutation in a high‐grade glioma originating from ganglioneuroma was treated with dabrafenib (a selective inhibitor of BRAF V600E) and TTFields after the failure of TMZ‐based therapy. More than 2 years of follow‐up have shown a complete response to the combination therapy.81 The superiority of gene‐targeted treatment in combination with TTFields therapy should be supported by the outcomes of large‐scale, multiply cohort clinical trials in the future.Metastatic tumorsThe majority of brain metastases result from lung cancer, while in women the most common primary histology is breast cancer.82 The development of brain metastases complicates many solid tumors and is attributed to the death of patients with advanced cancer.83 A METIS trial enrolling non‐small cells lung cancer (NSCLC) patients with 1–10 brain metastases (NCT02831959) who receive stereotactic radiosurgery (SRS) followed by TTFields (150 kHz, ≥18 h/day) or supportive care within 7 days of SRS reported no safety issues in 2018 and is ongoing currently.84PRECLINICAL STUDIES OF TTFIELDS IN MALIGNANT BRAIN TUMORSWith an optimal output pattern, TTFields could significantly inhibit the viability, proliferation, and invasiveness of different cell lines, irrespective of their genetic traits.85 Several studies have shown synergistic effects of TTFields and targeted agents, of which BEV is widely applied.68,86 Sorafenib is a multi‐kinase inhibitor and a first‐line agent for the treatment of high‐grade gliomas as well, but it has failed to improve the outcomes of patients when combined with TMZ.87 While combined with TTFields, sorafenib significantly inhibited motility, invasiveness, and angiogenesis of GBM cells.88 Evolutionary conserved protein kinase monopolar spindle 1 (MPS1) inhibition also has an impact on the mitotic process, resulting in more than an additive effect when combined with TTFields to GBM cells, and the anti‐proliferative benefit of the combination therapy begins to work earlier than mono‐therapy.89 Some inhibitors of MPS1 have been developed and can potentially serve as a bridge for TTFields therapy interruption. Likewise, hyperthermia has been reported to increase the efficacy of other approaches against cancers. Combining heat therapy with TTFields has been reported to enhance each other's therapeutic effects and inhibit the metastasis of GBM cells.90 The combination of two physical therapy may be easily tolerated by patients. Since Ca2+ channels contribute to cellular stress response to TTFields, combining TTFields with Ca2+ antagonists (e.g., benidipine) may augment the efficacy and outcome of TTFields.57 It provides the possibility of combining TTFields with Ca2+ antagonists, which are already applied in clinical. In brief, TTFields hold great promise to address the challenge across the spectrum of the management of patients with high‐grade gliomas by optimizing other treatment strategies.Given the special immune environment of intracranial tumors, brain malignancies suppress immune cell activity and anti‐cancer function. Revitalizing the central immune system has become the emerging for malignant brain tumors and experienced tremendous growth.5 Considering the transformation of tumors under TTFields exposure to a state more favorable for an anti‐tumor immune response,53 and its role of switching on the BBB, TTFields may potentially enhance the central anti‐tumor immune response.60,91 At present, immunotherapeutic approaches for GBM contain vaccines, checkpoint inhibitors, CAR‐T cells, and oncolytic viruses.5 Combining TTFields and anti‐PD‐1 (programmed cell death protein 1) therapy has already been proven more valid in the resistance to extra‐cranial tumors than single therapy by triggering immune response.50 Several clinical trials (e.g., NCT03223103, NCT03405792) to test the immunogenicity and safety of immunotherapeutic approaches in combination with TTFields in patients with malignant brain tumors are conducted currently, which will be valuable to establish a combination of therapeutic strategies and to elucidate the mechanism of TTFields on the immune micro‐environment of malignant brain tumors.THERAPEUTIC EVALUATION OF TTFIELDS THERAPYMagnetic resonance imaging (MRI) is the common method for diagnostics and follow‐up of malignant brain diseases, especially the change of MRI contrast enhancement, which is often considered an indicator of treatment response or tumor progression.92 But conventional MRI cannot assess treatment response reliably due to a lack of specificity,93 and the enhancement can be caused by other non‐tumor‐associated processes.94 Physiological imaging techniques, including diffusion tensor imaging (DTI), dynamic susceptibility contrast‐perfusion weighted imaging (DSC‐PWI) and proton MR spectroscopy, have great potential in assessing treatment response to different therapies in patients with GBM. A case reported the experience in evaluating treatment response to TTFields in combination with TMZ in a female ndGBM patient using DTI, DSC‐PWI and 3D‐echo‐planar spectroscopic imaging. Compared with baseline, increased mean diffusivity and decreased fractional anisotropy, maximum relative cerebral blood volume, and reduced choline/creatine were noted at 2 months follow‐up periods.95 The MRI results verified the synergistic effect of TTFields and TMZ chemotherapy can inhibit the growth of tumors.Positron emission tomography (PET) scanning provides more biological information than just anatomical information in a non‐invasive way.96 As the most widely clinically applied tracer, 18F‐FDG(18F‐2‐fluoro‐2‐deoxy‐D‐glucose) can be highly taken up by normal brain tissue, thus limiting the application in patients with malignant brain tumors.97 The Response Assessment in Neuro‐Oncology group has recommended more wide‐scale diagnostic access to amino acids‐based PET for the management of patients with malignant brain tumors,98 due to the low uptake by normal brain tissue.99 When FET (O‐(2‐18F‐fluoroethyl)‐L‐tyrosine) PET scanning was applied in patients with high‐grade glioma whose treatment included TTFields, the data showed increased tumor volume with increased uptake or metabolic activity and demonstrated with histologically or clinically follow‐up confirmed disease progression. Compared to MRI images, FET PET images present clearer lesion boundaries (Figure 2).100 AMT (alpha[C‐11]‐methyl‐L‐tryptophan) PET scanning has been proven to detect an early metabolic response in rGBM patients before and after TTFields therapy. The images indicated that AMT PET can detect the metabolic alterations of amino acids in GBM cells induced by TTFields earlier than MRI, therefore helping in clinical decision‐making, especially in cases where MRI images are inconclusive.101 18F‐DASA‐23 (1‐((2‐fluoro‐6‐18F‐fluorophenyl)sulfonyl)‐4‐((4‐methoxyphenyl)sulfonyl)piperazine) was introduced to provide the information on the level of pyruvate kinase M2 (PKM2),102 which is preferentially expressed in cancer cells and contributes to anabolic glucose metabolism.103 A research project on manipulating 18F‐DASA‐23 radiotracer to detect the impairment of GBM glycolytic metabolism through down‐regulation of the expression of PKM2 by TTFields was published.104 This research proved that TTFields can cause a shift in GBM metabolism from glycolysis to oxidative phosphorylation for the first time. Even in the presence of sufficient oxygen, cancer cells prefer to use the inefficient process of glycolysis for energy production,105 the so‐called Warburg effect, which produces lactic acid that is beneficial for tumor growth and metastasis.106 Warburg effect attributes to the tumor progression and provides a suitable and appropriate atmosphere for tumor to metastasize. Therefore, the metabolic reprogramming of tumor cells by TTFields may also be one of its significant anti‐cancer mechanisms.2FIGUREImages before TTFields and during follow‐up of a 73‐year‐old patient with GBM. Due to immune thrombocytopenia, only TTFields were applied 4 weeks after completion of radiotherapy with concomitant TMZ. Before TTFields, baseline FET PET images showed a slight increase in metabolic activity (TBRmax 2.0, TBRmean 1.6) without spatial counterpart contrast enhancement. Sequential FET PET images at 3 and 6 months follow‐up showed a decrease in metabolic activity, as evidenced by a decreased tumor‐to‐brain ratio. Compared to MRI T1 images, FET PET images showed clearer lesions. Adapted permission from (Ref. 100).TTFIELDS IMPROVE THE QUALITY OF PATIENT'S LIFETill now, accumulating clinical experience has confirmed the safety of TTFields therapy, and no significantly life‐threatening TTFields‐related event has been reported.11,12 The most frequent TTFields‐related side effect is skin reaction,107 while other therapies often cause serious systematic side effects. The array‐associated skin toxicity including rash, irritation, erythema, pruritus, contact dermatitis, ulceration, infection and skin breakdown, could be cured with topical corticosteroid creams, topical and oral antibiotics, and isolation of the affected skin surface from adhesives or pressure.108 In brief, TTFields are linked to prolonged survival outcomes, minimal adverse effects and are easily manageable.According to the patient registration dataset, the clinical efficacy of TTFields on patients was significantly improved with ≥75% daily adherence (≥18 h/day) compared with <75%.109 In other words, the efficacy of TTFields on patients with brain malignancies critically depends on their high compliance rate. Patients must abide by a long daily regimen of continuous TTFields for at least 4 weeks, which might be inconvenient for patients, allowing patients to receive treatment by simply wearing the devices on their heads and not interfering with their daily activities. The most commonly employed TTFields delivery system consists of four transducer arrays, a field generator, and a power resource. The distribution of the arrays can be determined by the NovoTAL software to ensure that the best field strength is obtained at the tumor site.110 EF is delivered through the transducer arrays attached to the skin, and patients with lesions in the brain need to make an adequate commitment to daily skin care, regular hair shaving, and carrying sufficient power sources during TTFields therapy. Patients are more likely to experience the psychological burden of changes in appearance, and the inconvenience of wearing the arrays and carrying the battery packs for several hours a day. Fortunately, according to the analysis of the EF‐14 trial, adding TTFields to cancer treatment does not worsen patient function and well‐being production, such as role, social, and physical functioning.107 Patients can enjoy symptomatically stability during TTFields therapy, meanwhile, their quality of life can be maintained.Since late 2019, the COVID‐19 epidemic has forced a change in the treatment paradigm brought about by quarantine limitations, which might become a barrier to the routine treatment of patients with cancer. TTFields can be administered via telemedicine and confer a competitive advantage to be conveniently employed for the management of GBM patients during the COVID‐19 pandemic.111 Furthermore, reducing patient visits to healthcare facilities and cutting down on potential exposure to the virus will ensure the safety of ongoing treatment.CONCLUSIONAs a noninvasive physical therapy, TTFields have exhibited unique advantages in the treatment of patients with malignant brain tumors and are well suited for clinical trials exploring combined approaches to the management of patients due to the minor adverse effects and absence of toxicity. Nevertheless, the application of TTFields in pediatric high‐grade glioma is relatively rare, with few case reports regarding safety analysis, and needs to be corroborated in a larger patient cohort.112 TTFields do not have a half‐life and enable sustainable treatment,113 which can be applied between chemoradiotherapy or concurrently with RT.114 Furthermore, TTFields are inherently non‐specific to tumor types, may cause therapeutic effects for a wide range of solid tumors, and are being investigated for application to extracranial tumors.50,63,115 Unfortunately, the mechanism of TTFields is far less clear and suffers from skepticism than other therapies.116 Pharmacological profiling may help reveal the synergistic effects of TTFields in combination with other therapeutic agents. Continued enhancement of research and understanding of the molecular mechanisms will facilitate the adoption of this novel therapy for integration into existing or new treatment strategies.Although already become an established treatment for ndGBM or rGBM in adults and has been proven effective in combination with SOC of GBM, TTFields were not recommended to be added to SOC of GBM by the leading experts.114 High cost is the biggest obstacle to broadening the application of TTFields, which needs to be addressed immediately, and additional cost‐effectiveness studies are recommended.117,118 Therefore, improving the efficiency of TTFields will greatly benefit cancer treatment and reduce global medical costs. The phase II clinical trial EF‐33 (NCT04492163) for rGBM is being carried out, which applies higher field strength to the tumor by increasing the number of electric field arrays under the condition of safety assurance to achieve a better therapeutic effect. To improve the convenience of use, further refinement of the TTFields system is required to improve the ease of use and device performance through physical and other means (e.g., changing the size and weight of the instrument, implanting electrodes around the tumor, and increasing battery capacity). Conducting an outpatient clinic for TTFields therapy consultation is suggested, which may lead to a great promotion of motivation and compliance rate, and ensures the clinical efficacy of TTFields.119 Besides, it's still a mission for researchers to figure out the biomarkers to select the suitable patients who might be responsive to TTFields, and the markers or technique predicting optimal frequency or response in different cancer types or individuals. The best combination formula based on TTFields for special populations should also be defined. The framework of TTFields therapy, from patient selection and treatment to the follow‐up efficacy assessment system, needs to be refined in detail. As an innovative treatment with great potential for tumor therapy, we firmly believe that the TTFields treatment system will be a boon to patients with cancer.AUTHOR CONTRIBUTIONSXiaoqing Xing: Writing – original draft (supporting). Jinyun Zhou: Writing – review and editing (lead). Han Jiang: Writing – review and editing (supporting). Peili Cen: Writing – review and editing (supporting). Chentao Jin: Writing – review and editing (supporting). Yan Zhong: Writing – review and editing (supporting). Rui Zhou: Writing – review and editing (equal). Hong Zhang: Conceptualization (equal); funding acquisition (equal); supervision (equal); validation (equal).ACKNOWLEDGMENTSNot applicable.FUNDING INFORMATIONThis work was supported by the National Natural Science Foundation of China (81725009, 21788102 and 82030049), and the National Key R&D Program of China (2021YFE0108300).CONFLICT OF INTEREST STATEMENTThe authors have stated explicitly that there are no conflicts of interest in connection with this articleDATA AVAILABILITY STATEMENTNot applicable.ETHICS STATEMENTNot applicable.REFERENCESOstrom Q, Gittleman H, Liao P, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007‐2011. Neuro Oncol. 2014;16:iv1‐iv63.Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz‐Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011–2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86.Davis M. Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420‐429.Han B, Meng X, Wu P, et al. ATRX/EZH2 complex epigenetically regulates FADD/PARP1 axis, contributing to TMZ resistance in glioma. Theranostics. 2020;10(7):3351‐3365.Ljubimov V, Ramesh A, Davani S, Danielpour M, Breunig J, Black K. Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID‐19 pandemic. Adv Drug Deliv Rev. 2022;181:114033.Rivoirard R, Vallard A, Langrand‐Escure J, et al. Thirty years of phase I radiochemotherapy trials: latest development. Eur J Cancer (Oxford, England: 1990). 2016;58:1‐7.Sun G, Rong D, Li Z, et al. Role of small molecule targeted compounds in cancer: Progress, opportunities, and challenges. Front Cell Dev Biol. 2021;9:694363.Lin X, DeAngelis LM. Treatment of brain metastases. J Clin Oncol. 2015;33(30):3475‐3484.Arvanitis CD, Ferraro GB, Jain RK. The blood‐brain barrier and blood‐tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41.Kirson E, Gurvich Z, Schneiderman R, et al. Disruption of cancer cell replication by alternating electric fields. Cancer Res. 2004;64(9):3288‐3295.Stupp R, Wong E, Kanner A, et al. NovoTTF‐100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer (Oxford, England: 1990). 2012;48(14):2192‐2202.Stupp R, Taillibert S, Kanner A, et al. Effect of tumor‐treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma: a randomized clinical trial. Jama. 2017;318(23):2306‐2316.Kutuk T, Appel H, Avendano M, et al. Feasibility of tumor treating fields with pemetrexed and platinum‐based chemotherapy for unresectable malignant pleural mesothelioma: single‐center, real‐world data. Cancer. 2022;14(8):2020.Robinson AJ, Jain A, Sherman HG, et al. Toward hijacking bioelectricity in cancer to develop new bioelectronic medicine. Adv Therapeut. 2021;4(3):2000248.Payne SL, Ram P, Srinivasan DH, Le TT, Levin M, Oudin MJ. Potassium channel‐driven bioelectric signalling regulates metastasis in triple‐negative breast cancer. EBioMedicine. 2022;75:103767.Levin M, Stevenson CG. Regulation of cell behavior and tissue patterning by bioelectrical signals: challenges and opportunities for biomedical engineering. Annu Rev Biomed Eng. 2012;14(1):295‐323.Siems LL, Kosman AJ, Osborne SL. A comparative study of short wave and microwave diathermy on blood flow; the role of the somatic and sympathetic nerves in the vascular response to deep tissue heating. Arch Phys Med Rehabil. 1948;29(12):759‐764.Palti Y. Stimulation of muscles and nerves by means of externally applied electrodes. Bull Res Counc Isr Sect E Exp Med. 1962;10:54‐56.Xu S, Luo C, Chen D, Tang L, Chen L, Liu Z. Whole transcriptome and proteome analyses identify potential targets and mechanisms underlying tumor treating fields against glioblastoma. Cell Death Dis. 2022;13(8):721.Gutin P, Wong E. Noninvasive application of alternating electric fields in glioblastoma: a fourth cancer treatment modality. American Society of Clinical Oncology Educational Book American Society of Clinical Oncology Annual Meeting 2012:126–131.Kline‐Smith S, Walczak C. Mitotic spindle assembly and chromosome segregation: refocusing on microtubule dynamics. Mol Cell. 2004;15(3):317‐327.Barreca M, Stathis A, Barraja P, Bertoni F. An overview on anti‐tubulin agents for the treatment of lymphoma patients. Pharmacol Ther. 2020;211:107552.Voloshin T, Schneiderman R, Volodin A, et al. Tumor treating fields (TTFields) hinder cancer cell motility through regulation of microtubule and acting dynamics. Cancer. 2020;12(10):3016.Tooley A, Gilden J, Jacobelli J, et al. Amoeboid T lymphocytes require the septin cytoskeleton for cortical integrity and persistent motility. Nat Cell Biol. 2009;11(1):17‐26.Gilden J, Peck S, Chen Y, Krummel M. The septin cytoskeleton facilitates membrane retraction during motility and blebbing. J Cell Biol. 2012;196(1):103‐114.Kirson E, Dbalý V, Tovarys F, et al. Alternating electric fields arrest cell proliferation in animal tumor models and human brain tumors. Proc Natl Acad Sci U S A. 2007;104(24):10152‐10157.Giladi M, Schneiderman R, Voloshin T, 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.Gera N, Yang A, Holtzman T, Lee S, Wong E, Swanson K. Tumor treating fields perturb the localization of septins and cause aberrant mitotic exit. PloS One. 2015;10(5):e0125269.Lee SX, Wong ET, Swanson KD. Mitosis interference of cancer cells during anaphase by electric field from NOVOTTF‐100A. Neuro Oncol. 2011;13:13‐14.Kim EH, Lee WS, Oh HK. Tumor‐treating fields in combination with sorafenib curtails the growth of colorectal carcinoma by inactivating AKT/STAT3 signaling. Transl Cancer Res. 2022;11(8):2553‐2561.Silginer M, Weller M, Stupp R, Roth P. Biological activity of tumor‐treating fields in preclinical glioma models. Cell Death Dis. 2017;8(4):e2753.Kim E, Kim Y, Song H, et al. Biological effect of an alternating electric field on cell proliferation and synergistic antimitotic effect in combination with ionizing radiation. Oncotarget. 2016;7(38):62267‐62279.Giladi M, Munster M, Schneiderman R, et al. Tumor treating fields (TTFields) delay DNA damage repair following radiation treatment of glioma cells. Radiat Oncol (London, England). 2017;12(1):206.Karanam N, Srinivasan K, Ding L, Sishc B, Saha D, Story M. Tumor‐treating fields elicit a conditional vulnerability to ionizing radiation via the downregulation of BRCA1 signaling and reduced DNA double‐strand break repair capacity in non‐small cell lung cancer cell lines. Cell Death Dis. 2017;8(3):e2711.Zhao W, Steinfeld J, Liang F, et al. BRCA1‐BARD1 promotes RAD51‐mediated homologous DNA pairing. Nature. 2017;550(7676):360‐365.Kais Z, Rondinelli B, Holmes A, et al. FANCD2 maintains fork stability in BRCA1/2‐deficient tumors and promotes alternative end‐joining DNA repair. Cell Rep. 2016;15(11):2488‐2499.Karanam N, Ding L, Aroumougame A, Story M. Tumor treating fields cause replication stress and interfere with DNA replication fork maintenance: implications for cancer therapy. Transl Res: J Lab Clin Med. 2020;217:33‐46.Kirson E, Giladi M, Gurvich Z, et al. Alternating electric fields (TTFields) inhibit metastatic spread of solid tumors to the lungs. Clin Exp Metastasis. 2009;26(7):633‐640.Shi P, Tian J, Ulm B, et al. Tumor treating fields suppression of Ciliogenesis enhances temozolomide toxicity. Front Oncol. 2022;12:837589.Kalluri R, Weinberg R. The basics of epithelial‐mesenchymal transition. J Clin Invest. 2009;119(6):1420‐1428.Kim E, Song H, Yoo S, Yoon M. Tumor treating fields inhibit glioblastoma cell migration, invasion and angiogenesis. Oncotarget. 2016;7(40):65125‐65136.Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2(3):161‐174.Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219.Inoue T, Nakayama Y, Li Y, et al. SIRT2 knockdown increases basal autophagy and prevents postslippage death by abnormally prolonging the mitotic arrest that is induced by microtubule inhibitors. FEBS J. 2014;281(11):2623‐2637.Kim E, Jo Y, Sai S, et al. Tumor‐treating fields induce autophagy by blocking the Akt2/miR29b axis in glioblastoma cells. Oncogene. 2019;38(39):6630‐6646.Holdgaard S, Cianfanelli V, Pupo E, et al. Selective autophagy maintains centrosome integrity and accurate mitosis by turnover of centriolar satellites. Nat Commun. 2019;10(1):4176.Tsujimoto Y, Shimizu S. Another way to die: autophagic programmed cell death. Cell Death Diff. 2005;12(2):1528‐1534.Shteingauz A, Porat Y, Voloshin T, et al. AMPK‐dependent autophagy upregulation serves as a survival mechanism in response to tumor treating fields (TTFields). Cell Death Dis. 2018;9(11):1074.Wong E, Lok E, Gautam S, Swanson K. Dexamethasone exerts profound immunologic interference on treatment efficacy for recurrent glioblastoma. Br J Cancer. 2015;113(2):232‐241.Voloshin T, Kaynan N, Davidi S, et al. Tumor‐treating fields (TTFields) induce immunogenic cell death resulting in enhanced antitumor efficacy when combined with anti‐PD‐1 therapy. Cancer Immunol Immunother: CII. 2020;69(7):1191‐1204.Martins I, Wang Y, Michaud M, et al. Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ. 2014;21(1):79‐91.Park J, Song K, Jung S, et al. Tumor‐treating fields induce RAW264.7 macrophage activation via NK‐κB/MAPK signaling pathways. Technol Cancer Res Treat. 2019;18:1533033819868225.Chen D, Le S, Hutchinson T, et al. Tumor treating fields dually activate STING and AIM2 inflammasomes to induce adjuvant immunity in glioblastoma. J Clin Invest. 2022;132:0021‐9738.Aguilar AA, Ho MC, Chang E, et al. Permeabilizing cell membranes with electric fields. Cancers (Basel). 2021;13(9):2283.Chang E, Patel C, Pohling C, et al. Tumor treating fields increases membrane permeability in glioblastoma cells. Cell Death Discov. 2018;4:113.Probst U, Fuhrmann I, Beyer L, Wiggermann P. Electrochemotherapy as a new modality in interventional oncology: a review. Technol Cancer Res Treat. 2018;17:1533033818785329.Neuhaus E, Zirjacks L, Ganser K, et al. Alternating electric fields (TTFields) activate Ca1.2 channels in human glioblastoma cells. Cancer. 2019;11(1):110.Chinigò G, Castel H, Chever O, Gkika D. TRP channels in brain tumors. Front Cell Dev Biol. 2021;9:617801.Li X, Yang F, Rubinsky B. A theoretical study on the biophysical mechanisms by which tumor treating fields affect tumor cells during mitosis. IEEE Trans Biomed Eng. 2020;67(9):2594‐2602.Kessler AF, Salvador E, Domrose D, et al. Blood brain barrier (BBB) integrity is affected by tumor treating fields (TTFields) in vitro and In vivo. Int J Radiat Oncol Biol Phys. 2019;105(1):S162‐S163.Salvador E, Köppl T, Hörmann J, et al. Tumor treating fields (TTFields) induce cell junction alterations in a human 3D In vitro model of the blood‐brain barrier. Pharmaceutics. 2023;15(1):185.Ceresoli G, Aerts J, Dziadziuszko R, et al. Tumour treating fields in combination with pemetrexed and cisplatin or carboplatin as first‐line treatment for unresectable malignant pleural mesothelioma (STELLAR): a multicentre, single‐arm phase 2 trial. Lancet Oncol. 2019;20(12):1702‐1709.Giladi M, Schneiderman R, Porat Y, et al. Mitotic disruption and reduced clonogenicity of pancreatic cancer cells in vitro and in vivo by tumor treating fields. Pancreatol: Off J Int Assoc Pancreatol (IAP). 2014;14(1):54‐63.Akbarnejad Z, Eskandary H, Dini L, et al. Cytotoxicity of temozolomide on human glioblastoma cells is enhanced by the concomitant exposure to an extremely low‐frequency electromagnetic field (100Hz, 100G). Biomed Pharmacother. 2017;92:254‐264.Branter J, Estevez‐Cebrero M, Diksin M, et al. Genome‐wide expression and anti‐proliferative effects of electric field therapy on pediatric and adult brain tumors. Int J Mol Sci. 2022;23(4):1982.Kirson ED, Wasserman Y, Izhaki A, et al. Modeling tumor growth kinetics and its implications for TTFields treatment planning. Neuro Oncol. 2010;12:48.Salzberg M, Kirson E, Palti Y, Rochlitz C. A pilot study with very low‐intensity, intermediate‐frequency electric fields in patients with locally advanced and/or metastatic solid tumors. Onkologie. 2008;31(7):362‐365.Lu G, Rao M, Zhu P, et al. Triple‐drug therapy with bevacizumab, irinotecan, and temozolomide plus tumor treating fields for recurrent glioblastoma: a retrospective study. Front Neurol. 2019;10:42.Korshoej A, Lukacova S, Lassen‐Ramshad Y, et al. OptimalTTF‐1: enhancing tumor treating fields therapy with skull remodeling surgery. A clinical phase I trial in adult recurrent glioblastoma. Neuro‐Oncol Adv. 2020;2(1):vdaa121.Lazaridis L, Bumes E, Cäcilia Spille D, et al. First multicentric real‐life experience with the combination of CCNU and temozolomide in newly diagnosed MGMT promoter methylated IDH wildtype glioblastoma. Neurooncol Adv. 2022;4(1):vdac137.Lazaridis L, Schäfer N, Teuber‐Hanselmann S, et al. Tumour treating fields (TTFields) in combination with lomustine and temozolomide in patients with newly diagnosed glioblastoma. J Cancer Res Clin Oncol. 2020;146(3):787‐792.Miller R, Song A, Ali A, et al. Scalp‐sparing radiation with concurrent temozolomide and tumor treating fields (SPARE) for patients with newly diagnosed glioblastoma. Front Oncol. 2022;12:896246.Bokstein F, Blumenthal D, Limon D, Harosh C, Ram Z, Grossman R. Concurrent tumor treating fields (TTFields) and radiation therapy for newly diagnosed glioblastoma: a prospective safety and feasibility study. Front Oncol. 2020;10:411.Norden A, Young G, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787.Iwamoto F, Abrey L, Beal K, et al. Patterns of relapse and prognosis after bevacizumab failure in recurrent glioblastoma. Neurology. 2009;73(15):1200‐1206.Rosen J, Blau T, Grau S, Barbe M, Fink G, Galldiks N. Extracranial metastases of a cerebral glioblastoma: a case report and review of the literature. Case Reports Oncol. 2018;11(2):591‐600.Mikic N, Poulsen F, Kristoffersen K, et al. Study protocol for OptimalTTF‐2: enhancing tumor treating fields with skull remodeling surgery for first recurrence glioblastoma: a phase 2, multi‐center, randomized, prospective, interventional trial. BMC Cancer. 2021;21(1):1010.Fisher J, Adamson D. Current FDA‐approved therapies for high‐grade malignant gliomas. Biomedicine. 2021;9(3):324.Lin Y, Chen B. Case report: tumor‐treating fields prolongs IDH‐mutant anaplastic astrocytoma progression‐free survival and pathological evolution to glioblastoma. Ann Transl Med. 2021;9(24):1804.Majores M, von Lehe M, Fassunke J, Schramm J, Becker AJ, Simon M. Tumor recurrence and malignant progression of gangliogliomas. Cancer. 2008;113(12):3355‐3363.Meletath S, Pavlick D, Brennan T, et al. Personalized treatment for a patient with a BRAF V600E mutation using dabrafenib and a tumor treatment fields device in a high‐grade glioma arising from ganglioglioma. J Nat Comprehens Cancer Network: JNCCN. 2016;14(11):1345‐1350.Achrol A, Rennert R, Anders C, et al. Brain metastases. Nat Rev Dis Prim. 2019;5(1):5.Kirschenbaum D, Amit I. Brain metastases: not all tumors are created equal. Neuron. 2022;110(7):1097‐1099.Mehta M, Gondi V, Ahluwalia M, Brown P. Radiosurgery followed by tumour treating fields (TTFields) for brain metastases (1‐10) from NSCLC in the phase III METIS trial. Ann Oncol. 2019;30:v659.Wu H, Yang L, Liu H, et al. Exploring the efficacy of tumor electric field therapy against glioblastoma: an in vivo and in vitro study. CNS Neurosci Ther. 2021;27(12):1587‐1604.Elzinga G, Wong E. Resolution of cystic enhancement to add‐on tumor treating electric fields for recurrent glioblastoma after incomplete response to bevacizumab. Case Reports Neurol. 2014;6(1):109‐115.Den R, Kamrava M, Sheng Z, et al. A phase I study of the combination of sorafenib with temozolomide and radiation therapy for the treatment of primary and recurrent high‐grade gliomas. Int J Radiat Oncol Biol Phys. 2013;85(2):321‐328.Jo Y, Kim E, Sai S, et al. Functional biological activity of sorafenib as a tumor‐treating field sensitizer for glioblastoma therapy. Int J Mol Sci. 2018;19(11):3684.Kessler A, Frömbling G, Gross F, et al. Effects of tumor treating fields (TTFields) on glioblastoma cells are augmented by mitotic checkpoint inhibition. Cell Death Discov. 2018;4:12.Jo Y, Han Y, Lee E, et al. The combination of tumor treating fields and hyperthermia has synergistic therapeutic effects in glioblastoma cells by downregulating STAT3. Am J Cancer Res. 2022;12(3):1423‐1432.Lee J, Grabowski M, Lathia J. Tumor treating fields: killing two birds with one stone. J Clin Invest. 2022;132(8):e159073.Wen P, Macdonald D, Reardon D, et al. Updated response assessment criteria for high‐grade gliomas: response assessment in neuro‐oncology working group. J Clin Oncol. 2010;28(11):1963‐1972.Jackson E, Barboriak D, Bidaut L, Meyer C. Magnetic resonance assessment of response to therapy: tumor change measurement, truth data and error sources. Transl Oncol. 2009;2(4):211‐215.Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent M. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453‐461.Mohan S, Chawla S, Wang S, et al. Assessment of early response to tumor‐treating fields in newly diagnosed glioblastoma using physiologic and metabolic MRI: initial experience. CNS Oncol. 2016;5(3):137‐144.Tian M, He X, Jin C, et al. Transpathology: molecular imaging‐based pathology. Eur J Nucl Med Mol Imaging. 2021;48(8):2338‐2350.Werner J, Lohmann P, Fink G, Langen K, Galldiks N. Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules (Basel, Switzerland). 2020;25(6):1471.Langen K, Watts C. Neuro‐oncology: amino acid PET for brain tumours – ready for the clinic? Nat Rev Neurol. 2016;12(7):375‐376.Singhal T, Narayanan T, Jain V, Mukherjee J, Mantil J. 11C‐L‐methionine positron emission tomography in the clinical management of cerebral gliomas. Mol Imaging Biol. 2008;10(1):1‐18.Ceccon G, Lazaridis L, Stoffels G, et al. Use of FET PET in glioblastoma patients undergoing neurooncological treatment including tumour‐treating fields: initial experience. Eur J Nucl Med Mol Imaging. 2018;45(9):1626‐1635.Bosnyák E, Barger G, Michelhaugh S, et al. Amino acid PET imaging of the early metabolic response during tumor‐treating fields (TTFields) therapy in recurrent glioblastoma. Clin Nucl Med. 2018;43(3):176‐179.Witney T, James M, Shen B, et al. PET imaging of tumor glycolysis downstream of hexokinase through noninvasive measurement of pyruvate kinase M2. Sci Transl Med. 2015;7(310):310ra169.Christofk H, Vander Heiden M, Harris M, et al. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452(7184):230‐233.Patel C, Beinat C, Xie Y, Chang E, Gambhir S. Tumor treating fields (TTFields) impairs aberrant glycolysis in glioblastoma as evaluated by [F]DASA‐23, a non‐invasive probe of pyruvate kinase M2 (PKM2) expression. Neoplasia (New York, NY). 2021;23(1):58‐67.DeBerardinis R, Chandel N. We need to talk about the Warburg effect. Nat Metab. 2020;2(2):127‐129.Xu K, Yin N, Peng M, et al. Glycolytic ATP fuels phosphoinositide 3‐kinase signaling to support effector T helper 17 cell responses. Immunity. 2021;54(5):976‐987.Taphoorn M, Dirven L, Kanner A, et al. Influence of treatment with tumor‐treating fields on health‐related quality of life of patients with newly diagnosed glioblastoma: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(4):495‐504.Lacouture M, Davis M, Elzinga G, et al. Characterization and management of dermatologic adverse events with the NovoTTF‐100A system, a novel anti‐mitotic electric field device for the treatment of recurrent glioblastoma. Semin Oncol. 2014;S1‐14:S1‐S14.Toms S, Kim C, Nicholas G, Ram Z. Increased compliance with tumor treating fields therapy is prognostic for improved survival in the treatment of glioblastoma: a subgroup analysis of the EF‐14 phase III trial. J Neurooncol. 2019;141(2):467‐473.Davies A, Weinberg U, Palti Y. Tumor treating fields: a new frontier in cancer therapy. Ann N Y Acad Sci. 2013;1291:86‐95.Gatson N, Barnholtz‐Sloan J, Drappatz J, et al. Tumor treating fields for glioblastoma therapy during the COVID‐19 pandemic. Front Oncol. 2021;11:679702.Green A, Mulcahy Levy J, Vibhakar R, et al. Tumor treating fields in pediatric high‐grade glioma. Child's Nerv Syst: ChNS: Off J Int Soc Pediat Neurosurg. 2017;33(7):1043‐1045.Wenger C, Miranda P, Salvador R, et al. A review on tumor‐treating fields (TTFields): clinical implications inferred from computational modeling. IEEE Rev Biomed Eng. 2018;11:195‐207.Mehta M, Wen P, Nishikawa R, Reardon D, Peters K. Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol. 2017;111:60‐65.Vergote I, von Moos R, Manso L, Van Nieuwenhuysen E, Concin N, Sessa C. Tumor treating fields in combination with paclitaxel in recurrent ovarian carcinoma: results of the INNOVATE pilot study. Gynecol Oncol. 2018;150(3):471‐477.Wick W. TTFields: where does all the skepticism come from? Neuro Oncol. 2016;18(3):303‐305.Burri S, Gondi V, Brown P, Mehta M. The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196.Bernard‐Arnoux F, Lamure M, Ducray F, Aulagner G, Honnorat J, Armoiry X. The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136.Haj A, Doenitz C, Hohenberger C, Brawanski A, Proescholdt MA. P01.144 TTFields for newly diagnosed glioblastoma: impact of consultation strategy. (1522–8517 (Print)).
Cancer Reports – Wiley
Published: May 1, 2023
Keywords: alternating electric field; cancer therapy; glioblastoma; malignant brain tumors; tumor treating fields
You can share this free article with as many people as you like with the url below! We hope you enjoy this feature!
Read and print from thousands of top scholarly journals.
Already have an account? Log in
Bookmark this article. You can see your Bookmarks on your DeepDyve Library.
To save an article, log in first, or sign up for a DeepDyve account if you don’t already have one.
Copy and paste the desired citation format or use the link below to download a file formatted for EndNote
Access the full text.
Sign up today, get DeepDyve free for 14 days.
All DeepDyve websites use cookies to improve your online experience. They were placed on your computer when you launched this website. You can change your cookie settings through your browser.