Skip to main content
Top
Published in: Cellular Oncology 6/2022

03-11-2022 | Glioma | Review

Role of nerves in neurofibromatosis type 1-related nervous system tumors

Authors: Ling-Ling Ge, Ming-Yan Xing, Hai-Bing Zhang, Qing-Feng Li, Zhi-Chao Wang

Published in: Cellular Oncology | Issue 6/2022

Login to get access

Abstract

Background

Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder that affects nearly 1 in 3000 infants. Neurofibromin inactivation and NF1 gene mutations are involved in various aspects of neuronal function regulation, including neuronal development induction, electrophysiological activity elevation, growth factor expression, and neurotransmitter release. NF1 patients often exhibit a predisposition to tumor development, especially in the nervous system, resulting in the frequent occurrence of peripheral nerve sheath tumors and gliomas. Recent evidence suggests that nerves play a role in the development of multiple tumor types, prompting researchers to investigate the nerve as a vital component in and regulator of the initiation and progression of NF1-related nervous system tumors.

Conclusion

In this review, we summarize existing evidence about the specific effects of NF1 mutation on neurons and emerging research on the role of nerves in neurological tumor development, promising a new set of selective and targeted therapies for NF1-related tumors.
Literature
2.
go back to reference J.M. Friedman, Epidemiology of neurofibromatosis type 1. Am. J. Med. Genet. 89, 1–6 (1999)CrossRef J.M. Friedman, Epidemiology of neurofibromatosis type 1. Am. J. Med. Genet. 89, 1–6 (1999)CrossRef
4.
go back to reference G.A. Mashour, P.H. Driever, M. Hartmann, S.N. Drissel, T. Zhang, B. Scharf, U. Felderhoff-Müser, S. Sakuma, R.E. Friedrich, R.L. Martuza, V.F. Mautner, A. Kurtz, Circulating growth factor levels are associated with tumorigenesis in neurofibromatosis type 1. Clin. cancer research: official J. Am. Association Cancer Res. 10, 5677–5683 (2004). https://doi.org/10.1158/1078-0432.Ccr-03-0769CrossRef G.A. Mashour, P.H. Driever, M. Hartmann, S.N. Drissel, T. Zhang, B. Scharf, U. Felderhoff-Müser, S. Sakuma, R.E. Friedrich, R.L. Martuza, V.F. Mautner, A. Kurtz, Circulating growth factor levels are associated with tumorigenesis in neurofibromatosis type 1. Clin. cancer research: official J. Am. Association Cancer Res. 10, 5677–5683 (2004). https://​doi.​org/​10.​1158/​1078-0432.​Ccr-03-0769CrossRef
9.
go back to reference G. Blanchard, M.P. Lafforgue, L. Lion-François, I. Kemlin, D. Rodriguez, P. Castelnau, M. Carneiro, P. Meyer, F. Rivier, S. Barbarot, Y. Chaix, Systematic MRI in NF1 children under six years of age for the diagnosis of optic pathway gliomas. Study and outcome of a French cohort. Eur. J. Pediatr. neurology: EJPN : official J. Eur. Pediatr. Neurol. Soc. 20, 275–281 (2016). https://doi.org/10.1016/j.ejpn.2015.12.002CrossRef G. Blanchard, M.P. Lafforgue, L. Lion-François, I. Kemlin, D. Rodriguez, P. Castelnau, M. Carneiro, P. Meyer, F. Rivier, S. Barbarot, Y. Chaix, Systematic MRI in NF1 children under six years of age for the diagnosis of optic pathway gliomas. Study and outcome of a French cohort. Eur. J. Pediatr. neurology: EJPN : official J. Eur. Pediatr. Neurol. Soc. 20, 275–281 (2016). https://​doi.​org/​10.​1016/​j.​ejpn.​2015.​12.​002CrossRef
11.
go back to reference F. D’Angelo, M. Ceccarelli, L. Tala, J. Garofano, V. Zhang, F.P. Frattini, G. Caruso, K.D. Lewis, L. Alfaro, G. Bauchet, D. Berzero, M. Cachia, L. Cangiano, J. Capelle, F. de Groot, F. DiMeco, W. Ducray, G. Farah, S. Finocchiaro, C. Goutagny, C. Kamiya-Matsuoka, H. Lavarino, V. Loiseau, C.E. Lorgis, I. Marras, D.H. McCutcheon, S. Nam, V. Ronchi, R. Saletti, J. Seizeur, M. Slopis, F. Suñol, P. Vandenbos, D. Varlet, C. Vidaud, V. Watts, D.E. Tabar, S.K. Reuss, D. Kim, K. Meyronet, H. Mokhtari, K.P. Salvador, M. Bhat, M. Eoli, A. Sanson, Lasorella, A. Iavarone, The molecular landscape of glioma in patients with Neurofibromatosis 1. Nat. Med. 25, 176–187 (2019). https://doi.org/10.1038/s41591-018-0263-8CrossRef F. D’Angelo, M. Ceccarelli, L. Tala, J. Garofano, V. Zhang, F.P. Frattini, G. Caruso, K.D. Lewis, L. Alfaro, G. Bauchet, D. Berzero, M. Cachia, L. Cangiano, J. Capelle, F. de Groot, F. DiMeco, W. Ducray, G. Farah, S. Finocchiaro, C. Goutagny, C. Kamiya-Matsuoka, H. Lavarino, V. Loiseau, C.E. Lorgis, I. Marras, D.H. McCutcheon, S. Nam, V. Ronchi, R. Saletti, J. Seizeur, M. Slopis, F. Suñol, P. Vandenbos, D. Varlet, C. Vidaud, V. Watts, D.E. Tabar, S.K. Reuss, D. Kim, K. Meyronet, H. Mokhtari, K.P. Salvador, M. Bhat, M. Eoli, A. Sanson, Lasorella, A. Iavarone, The molecular landscape of glioma in patients with Neurofibromatosis 1. Nat. Med. 25, 176–187 (2019). https://​doi.​org/​10.​1038/​s41591-018-0263-8CrossRef
13.
go back to reference S. Deborde, T. Omelchenko, A. Lyubchik, Y. Zhou, S. He, W.F. McNamara, N. Chernichenko, S.Y. Lee, F. Barajas, C.H. Chen, R.L. Bakst, E. Vakiani, S. He, A. Hall, R.J. Wong, Schwann cells induce cancer cell dispersion and invasion. J. Clin. Investig. 126, 1538–1554 (2016). https://doi.org/10.1172/jci82658CrossRef S. Deborde, T. Omelchenko, A. Lyubchik, Y. Zhou, S. He, W.F. McNamara, N. Chernichenko, S.Y. Lee, F. Barajas, C.H. Chen, R.L. Bakst, E. Vakiani, S. He, A. Hall, R.J. Wong, Schwann cells induce cancer cell dispersion and invasion. J. Clin. Investig. 126, 1538–1554 (2016). https://​doi.​org/​10.​1172/​jci82658CrossRef
30.
go back to reference T. Ozawa, N. Araki, S. Yunoue, H. Tokuo, L. Feng, S. Patrakitkomjorn, T. Hara, Y. Ichikawa, K. Matsumoto, K. Fujii, H. Saya, The neurofibromatosis type 1 gene product neurofibromin enhances cell motility by regulating actin filament dynamics via the Rho-ROCK-LIMK2-cofilin pathway. J. Biol. Chem. 280, 39524–39533 (2005). https://doi.org/10.1074/jbc.M503707200CrossRef T. Ozawa, N. Araki, S. Yunoue, H. Tokuo, L. Feng, S. Patrakitkomjorn, T. Hara, Y. Ichikawa, K. Matsumoto, K. Fujii, H. Saya, The neurofibromatosis type 1 gene product neurofibromin enhances cell motility by regulating actin filament dynamics via the Rho-ROCK-LIMK2-cofilin pathway. J. Biol. Chem. 280, 39524–39533 (2005). https://​doi.​org/​10.​1074/​jbc.​M503707200CrossRef
33.
go back to reference S. Patrakitkomjorn, D. Kobayashi, T. Morikawa, M.M. Wilson, N. Tsubota, A. Irie, T. Ozawa, M. Aoki, N. Arimura, K. Kaibuchi, H. Saya, N. Araki, Neurofibromatosis type 1 (NF1) tumor suppressor, neurofibromin, regulates the neuronal differentiation of PC12 cells via its associating protein, CRMP-2. J. Biol. Chem. 283, 9399–9413 (2008). https://doi.org/10.1074/jbc.M708206200CrossRef S. Patrakitkomjorn, D. Kobayashi, T. Morikawa, M.M. Wilson, N. Tsubota, A. Irie, T. Ozawa, M. Aoki, N. Arimura, K. Kaibuchi, H. Saya, N. Araki, Neurofibromatosis type 1 (NF1) tumor suppressor, neurofibromin, regulates the neuronal differentiation of PC12 cells via its associating protein, CRMP-2. J. Biol. Chem. 283, 9399–9413 (2008). https://​doi.​org/​10.​1074/​jbc.​M708206200CrossRef
39.
40.
go back to reference Y. Kawachi, H. Maruyama, Y. Ishitsuka, Y. Fujisawa, J. Furuta, Y. Nakamura, E. Ichikawa, M. Furumura, F. Otsuka, NF1 gene silencing induces upregulation of vascular endothelial growth factor expression in both Schwann and non-Schwann cells. Exp. Dermatol. 22, 262–265 (2013). https://doi.org/10.1111/exd.12115CrossRef Y. Kawachi, H. Maruyama, Y. Ishitsuka, Y. Fujisawa, J. Furuta, Y. Nakamura, E. Ichikawa, M. Furumura, F. Otsuka, NF1 gene silencing induces upregulation of vascular endothelial growth factor expression in both Schwann and non-Schwann cells. Exp. Dermatol. 22, 262–265 (2013). https://​doi.​org/​10.​1111/​exd.​12115CrossRef
41.
go back to reference T.A. Mitsiadis, M. Salmivirta, T. Muramatsu, H. Muramatsu, H. Rauvala, E. Lehtonen, M. Jalkanen, I. Thesleff, Expression of the heparin-binding cytokines, midkine (MK) and HB-GAM (pleiotrophin) is associated with epithelial-mesenchymal interactions during fetal development and organogenesis. Dev. (Cambridge England) 121, 37–51 (1995). https://doi.org/10.1242/dev.121.1.37CrossRef T.A. Mitsiadis, M. Salmivirta, T. Muramatsu, H. Muramatsu, H. Rauvala, E. Lehtonen, M. Jalkanen, I. Thesleff, Expression of the heparin-binding cytokines, midkine (MK) and HB-GAM (pleiotrophin) is associated with epithelial-mesenchymal interactions during fetal development and organogenesis. Dev. (Cambridge England) 121, 37–51 (1995). https://​doi.​org/​10.​1242/​dev.​121.​1.​37CrossRef
43.
go back to reference I. Atallah, A.M. Cieza Rivera, O.M. Rivero Lezcano, L. Tascón-González, C. González-Cortés, C. Diez, T. Tascón, Fernández-Villa, V. Martín, Increased serum concentrations of estrogen-induced growth factors Midkine and FGF2 in NF1 patients with plexiform neurofibroma. Am. J. translational Res. 14, 3180–3188 (2022) I. Atallah, A.M. Cieza Rivera, O.M. Rivero Lezcano, L. Tascón-González, C. González-Cortés, C. Diez, T. Tascón, Fernández-Villa, V. Martín, Increased serum concentrations of estrogen-induced growth factors Midkine and FGF2 in NF1 patients with plexiform neurofibroma. Am. J. translational Res. 14, 3180–3188 (2022)
50.
go back to reference P.Y. Martin, S. Doly, A.M. Hamieh, E. Chapuy, V. Canale, M. Drop, S. Chaumont-Dubel, X. Bantreil, F. Lamaty, A.J. Bojarski, P. Zajdel, A. Eschalier, P. Marin, C. Courteix, mTOR activation by constitutively active serotonin6 receptors as new paradigm in neuropathic pain and its treatment. Prog. Neurobiol. 193, 101846 (2020). https://doi.org/10.1016/j.pneurobio.2020.101846CrossRef P.Y. Martin, S. Doly, A.M. Hamieh, E. Chapuy, V. Canale, M. Drop, S. Chaumont-Dubel, X. Bantreil, F. Lamaty, A.J. Bojarski, P. Zajdel, A. Eschalier, P. Marin, C. Courteix, mTOR activation by constitutively active serotonin6 receptors as new paradigm in neuropathic pain and its treatment. Prog. Neurobiol. 193, 101846 (2020). https://​doi.​org/​10.​1016/​j.​pneurobio.​2020.​101846CrossRef
51.
go back to reference E. Doucet, K. Grychowska, P. Zajdel, J. Bockaert, P. Marin, C. Bécamel, Blockade of Serotonin 5-HT(6) Receptor Constitutive Activity Alleviates Cognitive Deficits in a Preclinical Model of Neurofibromatosis Type 1, Int. J. Mol. Sci. 22, (2021) https://doi.org/10.3390/ijms221810178 E. Doucet, K. Grychowska, P. Zajdel, J. Bockaert, P. Marin, C. Bécamel, Blockade of Serotonin 5-HT(6) Receptor Constitutive Activity Alleviates Cognitive Deficits in a Preclinical Model of Neurofibromatosis Type 1, Int. J. Mol. Sci. 22, (2021) https://​doi.​org/​10.​3390/​ijms221810178
57.
go back to reference I.R. Violante, M.J. Ribeiro, R.A. Edden, P. Guimarães, I. Bernardino, J. Rebola, G. Cunha, E. Silva, M. Castelo-Branco, GABA deficit in the visual cortex of patients with neurofibromatosis type 1: genotype-phenotype correlations and functional impact. Brain: a journal of neurology 136, 918–925 (2013). https://doi.org/10.1093/brain/aws368CrossRef I.R. Violante, M.J. Ribeiro, R.A. Edden, P. Guimarães, I. Bernardino, J. Rebola, G. Cunha, E. Silva, M. Castelo-Branco, GABA deficit in the visual cortex of patients with neurofibromatosis type 1: genotype-phenotype correlations and functional impact. Brain: a journal of neurology 136, 918–925 (2013). https://​doi.​org/​10.​1093/​brain/​aws368CrossRef
62.
70.
73.
74.
go back to reference H.S. Venkatesh, L.T. Tam, P.J. Woo, J. Lennon, S. Nagaraja, S.M. Gillespie, J. Ni, D.Y. Duveau, P.J. Morris, J.J. Zhao, C.J. Thomas, M. Monje, Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature 549, 533–537 (2017). https://doi.org/10.1038/nature24014CrossRef H.S. Venkatesh, L.T. Tam, P.J. Woo, J. Lennon, S. Nagaraja, S.M. Gillespie, J. Ni, D.Y. Duveau, P.J. Morris, J.J. Zhao, C.J. Thomas, M. Monje, Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature 549, 533–537 (2017). https://​doi.​org/​10.​1038/​nature24014CrossRef
75.
go back to reference H.S. Venkatesh, W. Morishita, A.C. Geraghty, D. Silverbush, S.M. Gillespie, M. Arzt, L.T. Tam, C. Espenel, A. Ponnuswami, L. Ni, P.J. Woo, K.R. Taylor, A. Agarwal, A. Regev, D. Brang, H. Vogel, S. Hervey-Jumper, D.E. Bergles, M.L. Suvà, R.C. Malenka, M. Monje, Electrical and synaptic integration of glioma into neural circuits. Nature 573, 539–545 (2019). https://doi.org/10.1038/s41586-019-1563-yCrossRef H.S. Venkatesh, W. Morishita, A.C. Geraghty, D. Silverbush, S.M. Gillespie, M. Arzt, L.T. Tam, C. Espenel, A. Ponnuswami, L. Ni, P.J. Woo, K.R. Taylor, A. Agarwal, A. Regev, D. Brang, H. Vogel, S. Hervey-Jumper, D.E. Bergles, M.L. Suvà, R.C. Malenka, M. Monje, Electrical and synaptic integration of glioma into neural circuits. Nature 573, 539–545 (2019). https://​doi.​org/​10.​1038/​s41586-019-1563-yCrossRef
76.
go back to reference V. Venkataramani, D.I. Tanev, C. Strahle, A. Studier-Fischer, L. Fankhauser, T. Kessler, C. Körber, M. Kardorff, M. Ratliff, R. Xie, H. Horstmann, M. Messer, S.P. Paik, J. Knabbe, F. Sahm, F.T. Kurz, A.A. Acikgöz, F. Herrmannsdörfer, A. Agarwal, D.E. Bergles, A. Chalmers, H. Miletic, S. Turcan, C. Mawrin, D. Hänggi, H.K. Liu, W. Wick, F. Winkler, T. Kuner, Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature 573, 532–538 (2019). https://doi.org/10.1038/s41586-019-1564-xCrossRef V. Venkataramani, D.I. Tanev, C. Strahle, A. Studier-Fischer, L. Fankhauser, T. Kessler, C. Körber, M. Kardorff, M. Ratliff, R. Xie, H. Horstmann, M. Messer, S.P. Paik, J. Knabbe, F. Sahm, F.T. Kurz, A.A. Acikgöz, F. Herrmannsdörfer, A. Agarwal, D.E. Bergles, A. Chalmers, H. Miletic, S. Turcan, C. Mawrin, D. Hänggi, H.K. Liu, W. Wick, F. Winkler, T. Kuner, Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature 573, 532–538 (2019). https://​doi.​org/​10.​1038/​s41586-019-1564-xCrossRef
77.
go back to reference A. Comba, S.M. Faisal, P.J. Dunn, A.E. Argento, T.C. Hollon, W.N. Al-Holou, M.L. Varela, D.B. Zamler, G.L. Quass, P.F. Apostolides, C. Abel 2nd, C.E. Brown, P.E. Kish, A. Kahana, C.G. Kleer, S. Motsch, M.G. Castro, P.R. Lowenstein, Spatiotemporal analysis of glioma heterogeneity reveals COL1A1 as an actionable target to disrupt tumor progression. Nat. Commun. 13, 3606 (2022). https://doi.org/10.1038/s41467-022-31340-1CrossRef A. Comba, S.M. Faisal, P.J. Dunn, A.E. Argento, T.C. Hollon, W.N. Al-Holou, M.L. Varela, D.B. Zamler, G.L. Quass, P.F. Apostolides, C. Abel 2nd, C.E. Brown, P.E. Kish, A. Kahana, C.G. Kleer, S. Motsch, M.G. Castro, P.R. Lowenstein, Spatiotemporal analysis of glioma heterogeneity reveals COL1A1 as an actionable target to disrupt tumor progression. Nat. Commun. 13, 3606 (2022). https://​doi.​org/​10.​1038/​s41467-022-31340-1CrossRef
78.
81.
82.
go back to reference J.C. Carlson, M. Cantu Gutierrez, B. Lozzi, E. Huang-Hobbs, W.D. Turner, B. Tepe, Y. Zhang, A.M. Herman, G. Rao, C.J. Creighton, J.D. Wythe, B. Deneen, Identification of diverse tumor endothelial cell populations in malignant glioma. Neuro-oncology 23, 932–944 (2021). https://doi.org/10.1093/neuonc/noaa297CrossRef J.C. Carlson, M. Cantu Gutierrez, B. Lozzi, E. Huang-Hobbs, W.D. Turner, B. Tepe, Y. Zhang, A.M. Herman, G. Rao, C.J. Creighton, J.D. Wythe, B. Deneen, Identification of diverse tumor endothelial cell populations in malignant glioma. Neuro-oncology 23, 932–944 (2021). https://​doi.​org/​10.​1093/​neuonc/​noaa297CrossRef
87.
go back to reference R. Liu, X.P. Qin, Y. Zhuang, Y. Zhang, H.B. Liao, J.C. Tang, M.X. Pan, F.F. Zeng, Y. Lei, R.X. Lei, S. Wang, A.C. Liu, J. Chen, Z.F. Zhang, D. Zhao, S.L. Wu, R.Z. Liu, Z.F. Wang, Q. Wan, Glioblastoma recurrence correlates with NLGN3 levels. Cancer Med. 7, 2848–2859 (2018). https://doi.org/10.1002/cam4.1538CrossRef R. Liu, X.P. Qin, Y. Zhuang, Y. Zhang, H.B. Liao, J.C. Tang, M.X. Pan, F.F. Zeng, Y. Lei, R.X. Lei, S. Wang, A.C. Liu, J. Chen, Z.F. Zhang, D. Zhao, S.L. Wu, R.Z. Liu, Z.F. Wang, Q. Wan, Glioblastoma recurrence correlates with NLGN3 levels. Cancer Med. 7, 2848–2859 (2018). https://​doi.​org/​10.​1002/​cam4.​1538CrossRef
92.
go back to reference K.L. Ligon, E. Huillard, S. Mehta, S. Kesari, H. Liu, J.A. Alberta, R.M. Bachoo, M. Kane, D.N. Louis, R.A. Depinho, D.J. Anderson, C.D. Stiles, D.H. Rowitch, Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma. Neuron 53, 503–517 (2007). https://doi.org/10.1016/j.neuron.2007.01.009CrossRef K.L. Ligon, E. Huillard, S. Mehta, S. Kesari, H. Liu, J.A. Alberta, R.M. Bachoo, M. Kane, D.N. Louis, R.A. Depinho, D.J. Anderson, C.D. Stiles, D.H. Rowitch, Olig2-regulated lineage-restricted pathway controls replication competence in neural stem cells and malignant glioma. Neuron 53, 503–517 (2007). https://​doi.​org/​10.​1016/​j.​neuron.​2007.​01.​009CrossRef
94.
go back to reference J. Li, S. Zhu, D. Kozono, K. Ng, D. Futalan, Y. Shen, J.C. Akers, T. Steed, D. Kushwaha, M. Schlabach, B.S. Carter, C.H. Kwon, F. Furnari, W. Cavenee, S. Elledge, C.C. Chen, Genome-wide shRNA screen revealed integrated mitogenic signaling between dopamine receptor D2 (DRD2) and epidermal growth factor receptor (EGFR) in glioblastoma. Oncotarget 5, 882–893 (2014). https://doi.org/10.18632/oncotarget.1801CrossRef J. Li, S. Zhu, D. Kozono, K. Ng, D. Futalan, Y. Shen, J.C. Akers, T. Steed, D. Kushwaha, M. Schlabach, B.S. Carter, C.H. Kwon, F. Furnari, W. Cavenee, S. Elledge, C.C. Chen, Genome-wide shRNA screen revealed integrated mitogenic signaling between dopamine receptor D2 (DRD2) and epidermal growth factor receptor (EGFR) in glioblastoma. Oncotarget 5, 882–893 (2014). https://​doi.​org/​10.​18632/​oncotarget.​1801CrossRef
96.
go back to reference A.S. Chi, R.S. Tarapore, M.D. Hall, N. Shonka, S. Gardner, Y. Umemura, A. Sumrall, Z. Khatib, S. Mueller, C. Kline, W. Zaky, S. Khatua, S.P. Weathers, Y. Odia, T.N. Niazi, D. Daghistani, I. Cherrick, D. Korones, M.A. Karajannis, X.T. Kong, J. Minturn, A. Waanders, I. Arillaga-Romany, T. Batchelor, P.Y. Wen, K. Merdinger, L. Schalop, M. Stogniew, J.E. Allen, W. Oster, M.P. Mehta, Pediatric and adult H3 K27M-mutant diffuse midline glioma treated with the selective DRD2 antagonist ONC201. J. Neurooncol. 145, 97–105 (2019). https://doi.org/10.1007/s11060-019-03271-3CrossRef A.S. Chi, R.S. Tarapore, M.D. Hall, N. Shonka, S. Gardner, Y. Umemura, A. Sumrall, Z. Khatib, S. Mueller, C. Kline, W. Zaky, S. Khatua, S.P. Weathers, Y. Odia, T.N. Niazi, D. Daghistani, I. Cherrick, D. Korones, M.A. Karajannis, X.T. Kong, J. Minturn, A. Waanders, I. Arillaga-Romany, T. Batchelor, P.Y. Wen, K. Merdinger, L. Schalop, M. Stogniew, J.E. Allen, W. Oster, M.P. Mehta, Pediatric and adult H3 K27M-mutant diffuse midline glioma treated with the selective DRD2 antagonist ONC201. J. Neurooncol. 145, 97–105 (2019). https://​doi.​org/​10.​1007/​s11060-019-03271-3CrossRef
97.
98.
go back to reference J.E. Allen, G. Krigsfeld, P.A. Mayes, L. Patel, D.T. Dicker, A.S. Patel, N.G. Dolloff, E. Messaris, K.A. Scata, W. Wang, J.Y. Zhou, G.S. Wu, and W.S. El-Deiry, Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci. Transl. Med. 5, 171ra117 (2013). https://doi.org/10.1126/scitranslmed.3004828CrossRef J.E. Allen, G. Krigsfeld, P.A. Mayes, L. Patel, D.T. Dicker, A.S. Patel, N.G. Dolloff, E. Messaris, K.A. Scata, W. Wang, J.Y. Zhou, G.S. Wu, and W.S. El-Deiry, Dual inactivation of Akt and ERK by TIC10 signals Foxo3a nuclear translocation, TRAIL gene induction, and potent antitumor effects. Sci. Transl. Med. 5, 171ra117 (2013). https://​doi.​org/​10.​1126/​scitranslmed.​3004828CrossRef
99.
go back to reference A. Arce-Sillas, E. Sevilla-Reyes, D.D. Álvarez-Luquín, A. Guevara-Salinas, M.C. Boll, C.A. Pérez-Correa, A.V. Vivas-Almazan, U. Rodríguez-Ortiz, C. Castellanos Barba, M. Hernandez, G. Fragoso, E. Sciutto, G. Cárdenas, and L.V. Adalid-Peralta, Expression of dopamine receptors in immune regulatory cells. Neuroimmunomodulation 26, 159–166 (2019). https://doi.org/10.1159/000501187CrossRef A. Arce-Sillas, E. Sevilla-Reyes, D.D. Álvarez-Luquín, A. Guevara-Salinas, M.C. Boll, C.A. Pérez-Correa, A.V. Vivas-Almazan, U. Rodríguez-Ortiz, C. Castellanos Barba, M. Hernandez, G. Fragoso, E. Sciutto, G. Cárdenas, and L.V. Adalid-Peralta, Expression of dopamine receptors in immune regulatory cells. Neuroimmunomodulation 26, 159–166 (2019). https://​doi.​org/​10.​1159/​000501187CrossRef
100.
102.
go back to reference K. Bhat, M. Saki, F. Cheng, L. He, L. Zhang, A. Ioannidis, D. Nathanson, J. Tsang, S.J. Bensinger, P.L. Nghiemphu, T.F. Cloughesy, L.M. Liau, H.I. Kornblum, F. Pajonk, Dopamine Receptor Antagonists, Radiation, and Cholesterol Biosynthesis in Mouse Models of Glioblastoma. J. Natl Cancer Inst. 113, 1094–1104 (2021). https://doi.org/10.1093/jnci/djab018CrossRef K. Bhat, M. Saki, F. Cheng, L. He, L. Zhang, A. Ioannidis, D. Nathanson, J. Tsang, S.J. Bensinger, P.L. Nghiemphu, T.F. Cloughesy, L.M. Liau, H.I. Kornblum, F. Pajonk, Dopamine Receptor Antagonists, Radiation, and Cholesterol Biosynthesis in Mouse Models of Glioblastoma. J. Natl Cancer Inst. 113, 1094–1104 (2021). https://​doi.​org/​10.​1093/​jnci/​djab018CrossRef
106.
go back to reference D. Hoyer, D.E. Clarke, J.R. Fozard, P.R. Hartig, G.R. Martin, E.J. Mylecharane, P.R. Saxena, P.P. Humphrey, International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (Serotonin). Pharmacol. Rev. 46, 157–203 (1994) D. Hoyer, D.E. Clarke, J.R. Fozard, P.R. Hartig, G.R. Martin, E.J. Mylecharane, P.R. Saxena, P.P. Humphrey, International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (Serotonin). Pharmacol. Rev. 46, 157–203 (1994)
109.
112.
go back to reference S. Otto-Meyer, R. DeFaccio, C. Dussold, E. Ladomersky, L. Zhai, K.L. Lauing, L.R. Bollu, C. Amidei, R.V. Lukas, D.M. Scholtens, D.A. Wainwright, A retrospective survival analysis of Glioblastoma patients treated with selective serotonin reuptake inhibitors. Brain Behav. Immun. Health 2, (2020) https://doi.org/10.1016/j.bbih.2019.100025 S. Otto-Meyer, R. DeFaccio, C. Dussold, E. Ladomersky, L. Zhai, K.L. Lauing, L.R. Bollu, C. Amidei, R.V. Lukas, D.M. Scholtens, D.A. Wainwright, A retrospective survival analysis of Glioblastoma patients treated with selective serotonin reuptake inhibitors. Brain Behav. Immun. Health 2, (2020) https://​doi.​org/​10.​1016/​j.​bbih.​2019.​100025
113.
go back to reference A. Jussofie, V. Reinhardt, R. Kalff, GABA binding sites: their density, their affinity to muscimol and their behaviour against neuroactive steroids in human gliomas of different degrees of malignancy. J. Neural Transm. Gen. Sections 96, 233–241 (1994). https://doi.org/10.1007/bf01294790CrossRef A. Jussofie, V. Reinhardt, R. Kalff, GABA binding sites: their density, their affinity to muscimol and their behaviour against neuroactive steroids in human gliomas of different degrees of malignancy. J. Neural Transm. Gen. Sections 96, 233–241 (1994). https://​doi.​org/​10.​1007/​bf01294790CrossRef
124.
go back to reference S. Ishiuchi, K. Tsuzuki, Y. Yoshida, N. Yamada, N. Hagimura, H. Okado, A. Miwa, H. Kurihara, Y. Nakazato, M. Tamura, T. Sasaki, S. Ozawa, Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat. Med. 8, 971–978 (2002). https://doi.org/10.1038/nm746CrossRef S. Ishiuchi, K. Tsuzuki, Y. Yoshida, N. Yamada, N. Hagimura, H. Okado, A. Miwa, H. Kurihara, Y. Nakazato, M. Tamura, T. Sasaki, S. Ozawa, Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat. Med. 8, 971–978 (2002). https://​doi.​org/​10.​1038/​nm746CrossRef
128.
go back to reference L. Iacovelli, A. Arcella, G. Battaglia, S. Pazzaglia, E. Aronica, P. Spinsanti, A. Caruso, E. De Smaele, A. Saran, A. Gulino, M. D’Onofrio, F. Giangaspero, F. Nicoletti, Pharmacological activation of mGlu4 metabotropic glutamate receptors inhibits the growth of medulloblastomas. J. neuroscience: official J. Soc. Neurosci. 26, 8388–8397 (2006). https://doi.org/10.1523/jneurosci.2285-06.2006CrossRef L. Iacovelli, A. Arcella, G. Battaglia, S. Pazzaglia, E. Aronica, P. Spinsanti, A. Caruso, E. De Smaele, A. Saran, A. Gulino, M. D’Onofrio, F. Giangaspero, F. Nicoletti, Pharmacological activation of mGlu4 metabotropic glutamate receptors inhibits the growth of medulloblastomas. J. neuroscience: official J. Soc. Neurosci. 26, 8388–8397 (2006). https://​doi.​org/​10.​1523/​jneurosci.​2285-06.​2006CrossRef
129.
go back to reference C. Zhang, X.R. Yuan, H.Y. Li, Z.J. Zhao, Y.W. Liao, X.Y. Wang, J. Su, S.S. Sang, Q. Liu, Anti-cancer effect of metabotropic glutamate receptor 1 inhibition in human glioma U87 cells: involvement of PI3K/Akt/mTOR pathway. Cell. Physiol. Biochem. 35, 419–432 (2015) https://doi.org/10.1159/000369707 C. Zhang, X.R. Yuan, H.Y. Li, Z.J. Zhao, Y.W. Liao, X.Y. Wang, J. Su, S.S. Sang, Q. Liu, Anti-cancer effect of metabotropic glutamate receptor 1 inhibition in human glioma U87 cells: involvement of PI3K/Akt/mTOR pathway. Cell. Physiol. Biochem. 35, 419–432 (2015) https://​doi.​org/​10.​1159/​000369707
137.
go back to reference A.C. Solga, W.W. Pong, K.Y. Kim, P.J. Cimino, J.A. Toonen, J. Walker, T. Wylie, V. Magrini, M. Griffith, O.L. Griffith, A. Ly, M.H. Ellisman, E.R. Mardis, D.H. Gutmann, RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia 17, 776–788 (2015) https://doi.org/10.1016/j.neo.2015.10.002 A.C. Solga, W.W. Pong, K.Y. Kim, P.J. Cimino, J.A. Toonen, J. Walker, T. Wylie, V. Magrini, M. Griffith, O.L. Griffith, A. Ly, M.H. Ellisman, E.R. Mardis, D.H. Gutmann, RNA Sequencing of Tumor-Associated Microglia Reveals Ccl5 as a Stromal Chemokine Critical for Neurofibromatosis-1 Glioma Growth. Neoplasia 17, 776–788 (2015) https://​doi.​org/​10.​1016/​j.​neo.​2015.​10.​002
138.
go back to reference C.Yu-Ju Wu, C.H. Chen, C.Y. Lin, L.Y. Feng, Y.C. Lin, K.C. Wei, C.Y. Huang, J.Y. Fang, P.Y. Chen, CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase 2. Neuro-oncology 22, 253–266 (2020). https://doi.org/10.1093/neuonc/noz189CrossRef C.Yu-Ju Wu, C.H. Chen, C.Y. Lin, L.Y. Feng, Y.C. Lin, K.C. Wei, C.Y. Huang, J.Y. Fang, P.Y. Chen, CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase 2. Neuro-oncology 22, 253–266 (2020). https://​doi.​org/​10.​1093/​neuonc/​noz189CrossRef
139.
go back to reference Y. Pan, M. Xiong, R. Chen, Y. Ma, C. Corman, M. Maricos, U. Kindler, M. Semtner, Y.H. Chen, S. Dahiya, D.H. Gutmann, Athymic mice reveal a requirement for T-cell-microglia interactions in establishing a microenvironment supportive of Nf1 low-grade glioma growth. Genes Dev. 32, 491–496 (2018). https://doi.org/10.1101/gad.310797.117CrossRef Y. Pan, M. Xiong, R. Chen, Y. Ma, C. Corman, M. Maricos, U. Kindler, M. Semtner, Y.H. Chen, S. Dahiya, D.H. Gutmann, Athymic mice reveal a requirement for T-cell-microglia interactions in establishing a microenvironment supportive of Nf1 low-grade glioma growth. Genes Dev. 32, 491–496 (2018). https://​doi.​org/​10.​1101/​gad.​310797.​117CrossRef
142.
go back to reference Y. Pan, J.D. Hysinger, T. Barron, N.F. Schindler, O. Cobb, X. Guo, B. Yalçın, C. Anastasaki, S.B. Mulinyawe, A. Ponnuswami, S. Scheaffer, Y. Ma, K.C. Chang, X. Xia, J.A. Toonen, J.J. Lennon, E.M. Gibson, J.R. Huguenard, L.M. Liau, J.L. Goldberg, M. Monje, D.H. Gutmann, NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature 594, 277–282 (2021). https://doi.org/10.1038/s41586-021-03580-6CrossRef Y. Pan, J.D. Hysinger, T. Barron, N.F. Schindler, O. Cobb, X. Guo, B. Yalçın, C. Anastasaki, S.B. Mulinyawe, A. Ponnuswami, S. Scheaffer, Y. Ma, K.C. Chang, X. Xia, J.A. Toonen, J.J. Lennon, E.M. Gibson, J.R. Huguenard, L.M. Liau, J.L. Goldberg, M. Monje, D.H. Gutmann, NF1 mutation drives neuronal activity-dependent initiation of optic glioma. Nature 594, 277–282 (2021). https://​doi.​org/​10.​1038/​s41586-021-03580-6CrossRef
146.
go back to reference S. Parrinello, L.A. Noon, M.C. Harrisingh, P. Wingfield Digby, L.H. Rosenberg, C.A. Cremona, P. Echave, A.M. Flanagan, L.F. Parada, A.C. Lloyd, NF1 loss disrupts Schwann cell-axonal interactions: a novel role for semaphorin 4F. Genes Dev. 22, 3335–3348 (2008). https://doi.org/10.1101/gad.490608CrossRef S. Parrinello, L.A. Noon, M.C. Harrisingh, P. Wingfield Digby, L.H. Rosenberg, C.A. Cremona, P. Echave, A.M. Flanagan, L.F. Parada, A.C. Lloyd, NF1 loss disrupts Schwann cell-axonal interactions: a novel role for semaphorin 4F. Genes Dev. 22, 3335–3348 (2008). https://​doi.​org/​10.​1101/​gad.​490608CrossRef
147.
go back to reference K.J. Radomska, F. Coulpier, A. Gresset, A. Schmitt, A. Debbiche, S. Lemoine, P. Wolkenstein, J.M. Vallat, P. Charnay, P. Topilko, Cellular Origin, Tumor Progression, and Pathogenic Mechanisms of Cutaneous Neurofibromas Revealed by Mice with Nf1 Knockout in Boundary Cap Cells. Cancer Discov. 9, 130–147 (2019). https://doi.org/10.1158/2159-8290.Cd-18-0156CrossRef K.J. Radomska, F. Coulpier, A. Gresset, A. Schmitt, A. Debbiche, S. Lemoine, P. Wolkenstein, J.M. Vallat, P. Charnay, P. Topilko, Cellular Origin, Tumor Progression, and Pathogenic Mechanisms of Cutaneous Neurofibromas Revealed by Mice with Nf1 Knockout in Boundary Cap Cells. Cancer Discov. 9, 130–147 (2019). https://​doi.​org/​10.​1158/​2159-8290.​Cd-18-0156CrossRef
148.
go back to reference F.L. Rice, G. Houk, J.P. Wymer, S.J.C. Gosline, J. Guinney, J. Wu, N. Ratner, M.P. Jankowski, S. La Rosa, M. Dockum, J.R. Storey, S.L. Carroll, P.J. Albrecht, V.M. Riccardi, The evolution and multi-molecular properties of NF1 cutaneous neurofibromas originating from C-fiber sensory endings and terminal Schwann cells at normal sites of sensory terminations in the skin. PloS one 14, e0216527 (2019) https://doi.org/10.1371/journal.pone.0216527 F.L. Rice, G. Houk, J.P. Wymer, S.J.C. Gosline, J. Guinney, J. Wu, N. Ratner, M.P. Jankowski, S. La Rosa, M. Dockum, J.R. Storey, S.L. Carroll, P.J. Albrecht, V.M. Riccardi, The evolution and multi-molecular properties of NF1 cutaneous neurofibromas originating from C-fiber sensory endings and terminal Schwann cells at normal sites of sensory terminations in the skin. PloS one 14, e0216527 (2019) https://​doi.​org/​10.​1371/​journal.​pone.​0216527
158.
go back to reference S.J. Kazmi, S.J. Byer, J.M. Eckert, A.N. Turk, R.P. Huijbregts, N.M. Brossier, W.E. Grizzle, F.M. Mikhail, K.A. Roth, S.L. Carroll, Transgenic mice overexpressing neuregulin-1 model neurofibroma-malignant peripheral nerve sheath tumor progression and implicate specific chromosomal copy number variations in tumorigenesis. Am. J. Pathol. 182, 646–667 (2013). https://doi.org/10.1016/j.ajpath.2012.11.017CrossRef S.J. Kazmi, S.J. Byer, J.M. Eckert, A.N. Turk, R.P. Huijbregts, N.M. Brossier, W.E. Grizzle, F.M. Mikhail, K.A. Roth, S.L. Carroll, Transgenic mice overexpressing neuregulin-1 model neurofibroma-malignant peripheral nerve sheath tumor progression and implicate specific chromosomal copy number variations in tumorigenesis. Am. J. Pathol. 182, 646–667 (2013). https://​doi.​org/​10.​1016/​j.​ajpath.​2012.​11.​017CrossRef
159.
go back to reference S.N. Brosius, A.N. Turk, S.J. Byer, N.M. Brossier, L. Kohli, A. Whitmire, F.M. Mikhail, K.A. Roth, S.L. Carroll, Neuregulin-1 overexpression and Trp53 haploinsufficiency cooperatively promote de novo malignant peripheral nerve sheath tumor pathogenesis. Acta Neuropathol. 127, 573–591 (2014). https://doi.org/10.1007/s00401-013-1209-3CrossRef S.N. Brosius, A.N. Turk, S.J. Byer, N.M. Brossier, L. Kohli, A. Whitmire, F.M. Mikhail, K.A. Roth, S.L. Carroll, Neuregulin-1 overexpression and Trp53 haploinsufficiency cooperatively promote de novo malignant peripheral nerve sheath tumor pathogenesis. Acta Neuropathol. 127, 573–591 (2014). https://​doi.​org/​10.​1007/​s00401-013-1209-3CrossRef
166.
go back to reference F.C. Yang, D.A. Ingram, S. Chen, Y. Zhu, J. Yuan, X. Li, X. Yang, S. Knowles, W. Horn, Y. Li, S. Zhang, Y. Yang, S.T. Vakili, M. Yu, D. Burns, K. Robertson, G. Hutchins, L.F. Parada, D.W. Clapp, Nf1-dependent tumors require a microenvironment containing Nf1+/-- and c-kit-dependent bone marrow. Cell 135, 437–448 (2008). https://doi.org/10.1016/j.cell.2008.08.041CrossRef F.C. Yang, D.A. Ingram, S. Chen, Y. Zhu, J. Yuan, X. Li, X. Yang, S. Knowles, W. Horn, Y. Li, S. Zhang, Y. Yang, S.T. Vakili, M. Yu, D. Burns, K. Robertson, G. Hutchins, L.F. Parada, D.W. Clapp, Nf1-dependent tumors require a microenvironment containing Nf1+/-- and c-kit-dependent bone marrow. Cell 135, 437–448 (2008). https://​doi.​org/​10.​1016/​j.​cell.​2008.​08.​041CrossRef
169.
go back to reference V. Pinna, V. Lanari, P. Daniele, F. Consoli, E. Agolini, K. Margiotti, I. Bottillo, I. Torrente, A. Bruselles, C. Fusilli, A. Ficcadenti, S. Bargiacchi, E. Trevisson, M. Forzan, S. Giustini, C. Leoni, G. Zampino, M.C. Digilio, B. Dallapiccola, M. Clementi, M. Tartaglia, A. De Luca, p.Arg1809Cys substitution in neurofibromin is associated with a distinctive NF1 phenotype without neurofibromas. Eur. J. Hum. genetics: EJHG 23, 1068–1071 (2015). https://doi.org/10.1038/ejhg.2014.243CrossRef V. Pinna, V. Lanari, P. Daniele, F. Consoli, E. Agolini, K. Margiotti, I. Bottillo, I. Torrente, A. Bruselles, C. Fusilli, A. Ficcadenti, S. Bargiacchi, E. Trevisson, M. Forzan, S. Giustini, C. Leoni, G. Zampino, M.C. Digilio, B. Dallapiccola, M. Clementi, M. Tartaglia, A. De Luca, p.Arg1809Cys substitution in neurofibromin is associated with a distinctive NF1 phenotype without neurofibromas. Eur. J. Hum. genetics: EJHG 23, 1068–1071 (2015). https://​doi.​org/​10.​1038/​ejhg.​2014.​243CrossRef
170.
go back to reference M. Upadhyaya, S.M. Huson, M. Davies, N. Thomas, N. Chuzhanova, S. Giovannini, D.G. Evans, E. Howard, B. Kerr, S. Griffiths, C. Consoli, L. Side, D. Adams, M. Pierpont, R. Hachen, A. Barnicoat, H. Li, P. Wallace, J.P. Van Biervliet, D. Stevenson, D. Viskochil, D. Baralle, E. Haan, V. Riccardi, P. Turnpenny, C. Lazaro, L. Messiaen, An absence of cutaneous neurofibromas associated with a 3-bp inframe deletion in exon 17 of the NF1 gene (c.2970–2972 delAAT): evidence of a clinically significant NF1 genotype-phenotype correlation. Am. J. Hum. Genet. 80, 140–151 (2007). https://doi.org/10.1086/510781CrossRef M. Upadhyaya, S.M. Huson, M. Davies, N. Thomas, N. Chuzhanova, S. Giovannini, D.G. Evans, E. Howard, B. Kerr, S. Griffiths, C. Consoli, L. Side, D. Adams, M. Pierpont, R. Hachen, A. Barnicoat, H. Li, P. Wallace, J.P. Van Biervliet, D. Stevenson, D. Viskochil, D. Baralle, E. Haan, V. Riccardi, P. Turnpenny, C. Lazaro, L. Messiaen, An absence of cutaneous neurofibromas associated with a 3-bp inframe deletion in exon 17 of the NF1 gene (c.2970–2972 delAAT): evidence of a clinically significant NF1 genotype-phenotype correlation. Am. J. Hum. Genet. 80, 140–151 (2007). https://​doi.​org/​10.​1086/​510781CrossRef
172.
go back to reference M. Scala, I. Schiavetti, F. Madia, C. Chelleri, G. Piccolo, A. Accogli, A. Riva, V. Salpietro, R. Bocciardi, G. Morcaldi, M. Di Duca, F. Caroli, A. Verrico, C. Milanaccio, G. Viglizzo, M. Traverso, S. Baldassari, P. Scudieri, M. Iacomino, G. Piatelli, C. Minetti, P. Striano, M.L. Garrè, P. De Marco, M.C. Diana, V. Capra, M. Pavanello, F. Zara, Genotype-Phenotype Correlations in Neurofibromatosis Type 1: A Single-Center Cohort Study. Cancers 13 (2021) https://doi.org/10.3390/cancers13081879 M. Scala, I. Schiavetti, F. Madia, C. Chelleri, G. Piccolo, A. Accogli, A. Riva, V. Salpietro, R. Bocciardi, G. Morcaldi, M. Di Duca, F. Caroli, A. Verrico, C. Milanaccio, G. Viglizzo, M. Traverso, S. Baldassari, P. Scudieri, M. Iacomino, G. Piatelli, C. Minetti, P. Striano, M.L. Garrè, P. De Marco, M.C. Diana, V. Capra, M. Pavanello, F. Zara, Genotype-Phenotype Correlations in Neurofibromatosis Type 1: A Single-Center Cohort Study. Cancers 13 (2021) https://​doi.​org/​10.​3390/​cancers13081879
177.
go back to reference S. Friedman, R. Levy, W. Garrett, D. Doval, Bondarde, S.J.C. Research, Clinical Benefit of INCB7839, a Potent and Selective Inhibitor of ADAM10 and ADAM17, in Combination with Trastuzumab in Metastatic HER2 Positive Breast Cancer Patients. Cancer Res 69, 5056–5056 (2009) S. Friedman, R. Levy, W. Garrett, D. Doval, Bondarde, S.J.C. Research, Clinical Benefit of INCB7839, a Potent and Selective Inhibitor of ADAM10 and ADAM17, in Combination with Trastuzumab in Metastatic HER2 Positive Breast Cancer Patients. Cancer Res 69, 5056–5056 (2009)
178.
go back to reference J. Infante, H.A. Burris, N. Lewis, A multicenter phase Ib study of the safety, pharmacokinetics, biological activity and clinical efficacy of INCB7839, a potent and selective inhibitor of ADAM10 and ADAM17. (2007) J. Infante, H.A. Burris, N. Lewis, A multicenter phase Ib study of the safety, pharmacokinetics, biological activity and clinical efficacy of INCB7839, a potent and selective inhibitor of ADAM10 and ADAM17. (2007)
179.
go back to reference M.W. Ronellenfitsch, P.N. Harter, M. Kirchner, C. Heining, B. Hutter, L. Gieldon, J. Schittenhelm, M.U. Schuhmann, M. Tatagiba, G. Marquardt, M. Wagner, V. Endris, C.H. Brandts, V.F. Mautner, E. Schröck, W. Weichert, B. Brors, A. von Deimling, M. Mittelbronn, J.P. Steinbach, D.E. Reuss, H. Glimm, A. Stenzinger, S. Fröhling, Targetable ERBB2 mutations identified in neurofibroma/schwannoma hybrid nerve sheath tumors. J. Clin. Investig. 130, 2488–2495 (2020). https://doi.org/10.1172/jci130787CrossRef M.W. Ronellenfitsch, P.N. Harter, M. Kirchner, C. Heining, B. Hutter, L. Gieldon, J. Schittenhelm, M.U. Schuhmann, M. Tatagiba, G. Marquardt, M. Wagner, V. Endris, C.H. Brandts, V.F. Mautner, E. Schröck, W. Weichert, B. Brors, A. von Deimling, M. Mittelbronn, J.P. Steinbach, D.E. Reuss, H. Glimm, A. Stenzinger, S. Fröhling, Targetable ERBB2 mutations identified in neurofibroma/schwannoma hybrid nerve sheath tumors. J. Clin. Investig. 130, 2488–2495 (2020). https://​doi.​org/​10.​1172/​jci130787CrossRef
180.
go back to reference B. Weiss, B.C. Widemann, P. Wolters, E. Dombi, A. Vinks, A. Cantor, J. Perentesis, E. Schorry, N. Ullrich, D.H. Gutmann, J. Tonsgard, D. Viskochil, B. Korf, R.J. Packer, M.J. Fisher, Sirolimus for progressive neurofibromatosis type 1-associated plexiform neurofibromas: a neurofibromatosis Clinical Trials Consortium phase II study. Neuro-oncology 17, 596–603 (2015). https://doi.org/10.1093/neuonc/nou235CrossRef B. Weiss, B.C. Widemann, P. Wolters, E. Dombi, A. Vinks, A. Cantor, J. Perentesis, E. Schorry, N. Ullrich, D.H. Gutmann, J. Tonsgard, D. Viskochil, B. Korf, R.J. Packer, M.J. Fisher, Sirolimus for progressive neurofibromatosis type 1-associated plexiform neurofibromas: a neurofibromatosis Clinical Trials Consortium phase II study. Neuro-oncology 17, 596–603 (2015). https://​doi.​org/​10.​1093/​neuonc/​nou235CrossRef
181.
go back to reference N.J. Ullrich, S.P. Prabhu, A.T. Reddy, M.J. Fisher, R. Packer, S. Goldman, N.J. Robison, D.H. Gutmann, D.H. Viskochil, J.C. Allen, B. Korf, A. Cantor, G. Cutter, C. Thomas, J.P. Perentesis, T. Mizuno, A.A. Vinks, P.E. Manley, S.N. Chi, M.W. Kieran, A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1-associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study. Neuro-oncology 22, 1527–1535 (2020). https://doi.org/10.1093/neuonc/noaa071CrossRef N.J. Ullrich, S.P. Prabhu, A.T. Reddy, M.J. Fisher, R. Packer, S. Goldman, N.J. Robison, D.H. Gutmann, D.H. Viskochil, J.C. Allen, B. Korf, A. Cantor, G. Cutter, C. Thomas, J.P. Perentesis, T. Mizuno, A.A. Vinks, P.E. Manley, S.N. Chi, M.W. Kieran, A phase II study of continuous oral mTOR inhibitor everolimus for recurrent, radiographic-progressive neurofibromatosis type 1-associated pediatric low-grade glioma: a Neurofibromatosis Clinical Trials Consortium study. Neuro-oncology 22, 1527–1535 (2020). https://​doi.​org/​10.​1093/​neuonc/​noaa071CrossRef
Metadata
Title
Role of nerves in neurofibromatosis type 1-related nervous system tumors
Authors
Ling-Ling Ge
Ming-Yan Xing
Hai-Bing Zhang
Qing-Feng Li
Zhi-Chao Wang
Publication date
03-11-2022
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 6/2022
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-022-00723-3

Other articles of this Issue 6/2022

Cellular Oncology 6/2022 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine