Pilot study evaluating everolimus molecular mechanisms in tuberous sclerosis complex and focal cortical dysplasia

Dominique F Leitner, Evgeny Kanshin, Manor Askenazi, Yik Siu, Daniel Friedman, Sasha Devore, Drew Jones, Beatrix Ueberheide, Thomas Wisniewski, Orrin Devinsky, Dominique F Leitner, Evgeny Kanshin, Manor Askenazi, Yik Siu, Daniel Friedman, Sasha Devore, Drew Jones, Beatrix Ueberheide, Thomas Wisniewski, Orrin Devinsky

Abstract

Background: Tuberous sclerosis complex (TSC) and some focal cortical dysplasias (FCDs) are associated with dysfunctional mTOR signaling, resulting in increased cell growth and ribosomal S6 protein phosphorylation (phospho-S6). mTOR inhibitors can reduce TSC tumor growth and seizure frequency, and preclinical FCD studies indicate seizure suppression. This pilot study evaluated safety of mTOR inhibitor everolimus in treatment resistant (failure of >2 anti-seizure medications) TSC and FCD patients undergoing surgical resection and to assess mTOR signaling and molecular pathways.

Methods and findings: We evaluated everolimus in 14 treatment resistant epilepsy patients undergoing surgical resection (4.5 mg/m2 daily for 7 days; n = 4 Active, mean age 18.3 years, range 4-26; n = 10, Control, mean age 13.1, range 3-45). Everolimus was well tolerated. Mean plasma everolimus in Active participants were in target range (12.4 ng/ml). Brain phospho-S6 was similar in Active and Control participants with a lower trend in Active participants, with Ser235/236 1.19-fold (p = 0.67) and Ser240/244 1.15-fold lower (p = 0.66). Histologically, Ser235/236 was 1.56-fold (p = 0.37) and Ser240/244 was 5.55-fold lower (p = 0.22). Brain proteomics identified 11 proteins at <15% false discovery rate associated with coagulation system (p = 1.45x10-9) and acute phase response (p = 1.23x10-6) activation. A weighted gene correlation network analysis (WGCNA) of brain proteomics and phospho-S6 identified 5 significant modules. Higher phospho-S6 correlated negatively with cellular respiration and synaptic transmission and positively with organophosphate metabolic process, nuclear mRNA catabolic process, and neuron ensheathment. Brain metabolomics identified 14 increased features in Active participants, including N-acetylaspartylglutamic acid. Plasma proteomics and cytokine analyses revealed no differences.

Conclusions: Short-term everolimus before epilepsy surgery in TSC and FCD resulted in no adverse events and trending lower mTOR signaling (phospho-S6). Future studies should evaluate implications of our findings, including coagulation system activation and everolimus efficacy in FCD, in larger studies with long-term treatment to better understand molecular and clinical effects.

Clinical trials registration: ClinicalTrials.gov NCT02451696.

Conflict of interest statement

I have read the journal’s policy and the authors of this manuscript have the following competing interests: All the authors report no conflicts of interest. Below are the disclosures of the three authors: Daniel Friedman receives salary support for consulting and clinical trial related activities performed on behalf of The Epilepsy Study Consortium, a non-profit organization. Dr. Friedman receives no personal income for these activities. NYU receives a fixed amount from the Epilepsy Study Consortium towards Dr. Friedman’s salary. Within the past two years, The Epilepsy Study Consortium received payments for research services performed by Dr. Friedman from: Alterity, Baergic, Biogen, BioXcell, Cerevel, Cerebral, Jannsen, Lundbeck, Neurocrine, SK Life Science, and Xenon. He has also served as a paid consultant for Neurelis Pharmaceuticals and Receptor Life Sciences. He has received research support from NINDS, CDC, Epitel, and Neuropace unrelated to this study. He holds equity interests in Neuroview Technology. He received royalty income from Oxford University Press. Sasha Devore receives salary support from the National Institutes of Health, Department of Defense, and the Templeton World Charity Foundation unrelated to this study. Orrin Devinsky receives grant support from NINDS, NIMH, MURI, CDC and NSF. He has equity and/or compensation from the following companies: Tilray, Receptor Life Sciences, Qstate Biosciences, Tevard, Empatica, Engage, Egg Rock/Papa & Barkley, Rettco, SilverSpike, and California Cannabis Enterprises (CCE). He has received consulting fees from Zogenix, Ultragenyx, BridgeBio, and Marinus. He holds patents for the use of cannabidiol in treating neurological disorders but these are owned by GW Pharmaceuticals and he has waived any financial interests. He holds other patents in molecular biology. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Ribosomal S6 phosphorylation in whole…
Fig 1. Ribosomal S6 phosphorylation in whole brain homogenate.
A) Representative western blot images from surgically resected brain tissue of Active participants after taking 4.5 mg/m2 everolimus for 7 days and controls. Total S6, phospho-S6 (Ser235/236), phospho-S6 (Ser240/244), and beta-actin as loading control were evaluated in 14 participants. TSC participants are indicated “*”, while all others are FCD participants. B) Quantification of total S6 relative to actin indicates variability in baseline levels from sampled brain tissue, as well as one patient with increased expression of a smaller detected band (NYU-002). C) Percentage of phospho-S6 (Ser235/236) relative to total S6 indicates an average 1.19-fold decrease when comparing all Active participants to controls (p = 0.67). The highest level was seen in NYU-004. D) Percentage of phospho-S6 (Ser240/244) relative to total S6 indicates an average 1.15-fold decrease when comparing all Active participants to controls (p = 0.66). The highest level was seen in NYU-004. Error bars indicate SEM.
Fig 2. Histological quantification of ribosomal S6…
Fig 2. Histological quantification of ribosomal S6 phosphorylation in gray matter.
Representative images show total S6, phospho-S6 (Ser235/236), and phospho-S6 (Ser240/244) (red) in resected cortical tissue from FCD participants that were Controls A-C) or Active D-F) and in TSC participants that were Controls G-I) or Active J-L). M) Quantification of total S6 expression in the gray matter indicated no difference in baseline expression between Active and Control participants. N) Quantification of percent phospho-S6 (Ser235/236) relative to total S6 in the gray matter indicated an average 1.56-fold decrease when comparing all Active participants to Controls (p = 0.37). O) Quantification of percent phospho-S6 (Ser240/244) relative to total S6 in the gray matter indicated an average 5.55-fold decrease when comparing all Active participants to Controls (p = 0.22). Histology was performed on all cases with available FFPE, excluding 1 Active FCD case. Error bars indicate SEM. Scale bar represents 100 μm.
Fig 3. Proteomics in whole brain homogenate.
Fig 3. Proteomics in whole brain homogenate.
Proteomics was performed in surgically resected brain tissue from Active and Control participants. A) PCA in brain tissue indicated no segregation of Active and Control participants in PCA1 (p = 0.73) and with some segregation in PCA2 (p = 0.016). Clinical diagnoses (FCD, TSC) are noted as well. B) Differential expression analysis in brain tissue indicated that there were 11 significantly altered proteins in Active versus Control participants at an FDR < 15% (dotted line), detailed in S1 Table and S1 Fig. There were no significant proteins at FDR < 5–10%. C) WGCNA of brain proteomics and phospho-S6 evaluated in total brain homogenate by western blot, regardless of everolimus and clinical diagnoses. There were 1109 proteins that correlated with phospho-S6 levels (P235/236, p < 0.05), distributed across 10 modules (all but M-Purple). The top significantly correlated protein with P235/236 was a positive correlation to HSPA2 (p = 1.18 x 10−6, R2 = 0.87) in the M-Brown module. D) The top significantly correlated protein with P240/244 was a negative correlation with ANK2 (p = 7.92 x 10−5, R2 = 0.74) in the M-Brown module. E) Module trait correlation identified 5 significantly associated modules with phospho-S6 (p < 0.05). Modules were clustered by eigenprotein adjacency (relatedness to other modules) on the left. Name of module is indicated by “M-color.” P values are indicated for those modules with p < 0.05 correlation. Positive correlation is indicated in red and negative correlation in blue. Top module GO annotations are noted on the right (FDR < 5%, at least 5 proteins/annotation). Several modules did not have a significant GO annotation (“n.s.”).
Fig 4. Metabolomics in whole brain homogenate.
Fig 4. Metabolomics in whole brain homogenate.
Global polar metabolomics by LC-MS was performed on surgically resected brain tissue from Active and Control participants. A) PCA indicated distribution of participants, as well as clinical diagnoses (FCD, TSC). There was no segregation of Active and Control participants in PCA1 (p = 0.32) or PCA2 (p = 0.48). B) Differential expression analysis (p < 0.05, log2 (fold change) > 1) identified 14 metabolite features that were increased in Active participants, associated with the indicated annotations detailed in C).
Fig 5. Plasma proteomics and cytokine levels.
Fig 5. Plasma proteomics and cytokine levels.
A) Proteomics was performed on plasma obtained at time of surgical resection after 7 days everolimus. PCA indicates no segregation of Active from Control participants, nor by clinical diagnoses (FCD, TSC). B) Differential expression analysis of plasma proteins indicated no altered proteins in Active versus Control participants at FDR < 5%, nor at FDR < 15%. C-K) Plasma cytokine levels were evaluated at time of surgical resection. There were no significant differences in the cytokines evaluated.

References

    1. D’Gama AM, Poduri A. Precision Therapy for Epilepsy Related to Brain Malformations. Neurotherapeutics. 2021. Epub 20211004. doi: 10.1007/s13311-021-01122-6 .
    1. Goldstein HE, Hauptman JS. The Putative Role of mTOR Inhibitors in Non-tuberous Sclerosis Complex-Related Epilepsy. Front Neurol. 2021;12:639319. Epub 20210212. doi: 10.3389/fneur.2021.639319 .
    1. Mühlebner A, van Scheppingen J, Hulshof HM, Scholl T, Iyer AM, Anink JJ, et al.. Novel Histopathological Patterns in Cortical Tubers of Epilepsy Surgery Patients with Tuberous Sclerosis Complex. PLoS One. 2016;11(6):e0157396. Epub 20160613. doi: 10.1371/journal.pone.0157396 .
    1. Ellison D, Love S, Chimelli L, Harding B, Lowe J, Vinters H, et al.. Neuropathology: a reference text of CNS pathology. Third ed: MOSBY an imprint of Elsevier Limited; 2013.
    1. Blümcke I, Thom M, Aronica E, Armstrong DD, Vinters HV, Palmini A, et al.. The clinicopathologic spectrum of focal cortical dysplasias: a consensus classification proposed by an ad hoc Task Force of the ILAE Diagnostic Methods Commission. Epilepsia. 2011;52(1):158–74. Epub 2010/11/10. doi: 10.1111/j.1528-1167.2010.02777.x .
    1. Baldassari S, Ribierre T, Marsan E, Adle-Biassette H, Ferrand-Sorbets S, Bulteau C, et al.. Dissecting the genetic basis of focal cortical dysplasia: a large cohort study. Acta Neuropathol. 2019;138(6):885–900. Epub 2019/08/23. doi: 10.1007/s00401-019-02061-5 .
    1. Sim NS, Ko A, Kim WK, Kim SH, Kim JS, Shim KW, et al.. Precise detection of low-level somatic mutation in resected epilepsy brain tissue. Acta Neuropathol. 2019;138(6):901–12. Epub 20190803. doi: 10.1007/s00401-019-02052-6 .
    1. Leitner DF, Faustin A, Verducci C, Friedman D, William C, Devore S, et al.. Neuropathology in the North American sudden unexpected death in epilepsy registry. Brain Commun. 2021;3(3):fcab192. Epub 20210823. doi: 10.1093/braincomms/fcab192 .
    1. Klawitter J, Nashan B, Christians U. Everolimus and sirolimus in transplantation-related but different. Expert Opin Drug Saf. 2015;14(7):1055–70. Epub 20150426. doi: 10.1517/14740338.2015.1040388 .
    1. MacKeigan JP, Krueger DA. Differentiating the mTOR inhibitors everolimus and sirolimus in the treatment of tuberous sclerosis complex. Neuro Oncol. 2015;17(12):1550–9. Epub 20150819. doi: 10.1093/neuonc/nov152 .
    1. Granata S, Dalla Gassa A, Carraro A, Brunelli M, Stallone G, Lupo A, et al.. Sirolimus and Everolimus Pathway: Reviewing Candidate Genes Influencing Their Intracellular Effects. Int J Mol Sci. 2016;17(5). Epub 20160514. doi: 10.3390/ijms17050735 .
    1. Meikle L, Pollizzi K, Egnor A, Kramvis I, Lane H, Sahin M, et al.. Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function. J Neurosci. 2008;28(21):5422–32. doi: 10.1523/JNEUROSCI.0955-08.2008 .
    1. Fox JH, Connor T, Chopra V, Dorsey K, Kama JA, Bleckmann D, et al.. The mTOR kinase inhibitor Everolimus decreases S6 kinase phosphorylation but fails to reduce mutant huntingtin levels in brain and is not neuroprotective in the R6/2 mouse model of Huntington’s disease. Mol Neurodegener. 2010;5:26. Epub 20100622. doi: 10.1186/1750-1326-5-26 .
    1. Franz DN, Belousova E, Sparagana S, Bebin EM, Frost MD, Kuperman R, et al.. Long-Term Use of Everolimus in Patients with Tuberous Sclerosis Complex: Final Results from the EXIST-1 Study. PLoS One. 2016;11(6):e0158476. Epub 20160628. doi: 10.1371/journal.pone.0158476 .
    1. French JA, Lawson JA, Yapici Z, Ikeda H, Polster T, Nabbout R, et al.. Adjunctive everolimus therapy for treatment-resistant focal-onset seizures associated with tuberous sclerosis (EXIST-3): a phase 3, randomised, double-blind, placebo-controlled study. Lancet. 2016;388(10056):2153–63. Epub 20160906. doi: 10.1016/S0140-6736(16)31419-2 .
    1. Cloughesy TF, Yoshimoto K, Nghiemphu P, Brown K, Dang J, Zhu S, et al.. Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med. 2008;5(1):e8. doi: 10.1371/journal.pmed.0050008 .
    1. Northrup H, Krueger DA, Group ITSCC. Tuberous sclerosis complex diagnostic criteria update: recommendations of the 2012 Iinternational Tuberous Sclerosis Complex Consensus Conference. Pediatr Neurol. 2013;49(4):243–54. doi: 10.1016/j.pediatrneurol.2013.08.001 .
    1. Bruce J, Russell EM, Mollison J, Krukowski ZH. The measurement and monitoring of surgical adverse events. Health Technol Assess. 2001;5(22):1–194. doi: 10.3310/hta5220 .
    1. Chapter 57—Pharmacology and Molecular Mechanisms of Antineoplastic Agents for Hematologic Malignancies. In: Ronald Hoffman and Edward JBaLESaHEHaJIWaJAaMESaSAA, editor. Hematology (Seventh Edition). Seventh Edition ed: Elsevier; 2018. p. 849–912.
    1. Leitner DF, Mills JD, Pires G, Faustin A, Drummond E, Kanshin E, et al.. Proteomics and Transcriptomics of the Hippocampus and Cortex in SUDEP and High-Risk SUDEP Patients. Neurology. 2021. Epub 2021/04/28. doi: 10.1212/WNL.0000000000011999 .
    1. Zougman A, Selby PJ, Banks RE. Suspension trapping (STrap) sample preparation method for bottom-up proteomics analysis. Proteomics. 2014;14(9):1006–0. Epub 2014/03/29. doi: 10.1002/pmic.201300553 .
    1. Zecha J, Satpathy S, Kanashova T, Avanessian SC, Kane MH, Clauser KR, et al.. TMT Labeling for the Masses: A Robust and Cost-efficient, In-solution Labeling Approach. Mol Cell Proteomics. 2019;18(7):1468–78. Epub 2019/04/11. doi: 10.1074/mcp.TIR119.001385 .
    1. Bekker-Jensen DB, Kelstrup CD, Batth TS, Larsen SC, Haldrup C, Bramsen JB, et al.. An Optimized Shotgun Strategy for the Rapid Generation of Comprehensive Human Proteomes. Cell Syst. 2017;4(6):587–99 e4. Epub 2017/06/12. doi: 10.1016/j.cels.2017.05.009 .
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26(12):1367–72. Epub 2008/11/26. doi: 10.1038/nbt.1511 .
    1. Cox J, Neuhauser N, Michalski A, Scheltema RA, Olsen JV, Mann M. Andromeda: a peptide search engine integrated into the MaxQuant environment. J Proteome Res. 2011;10(4):1794–805. Epub 2011/01/25. doi: 10.1021/pr101065j .
    1. Cox J, Michalski A, Mann M. Software lock mass by two-dimensional minimization of peptide mass errors. J Am Soc Mass Spectrom. 2011;22(8):1373–80. Epub 2011/09/29. doi: 10.1007/s13361-011-0142-8 .
    1. Huang T, Choi M, Tzouros M, Golling S, Pandya NJ, Banfai B, et al.. MSstatsTMT: Statistical Detection of Differentially Abundant Proteins in Experiments with Isobaric Labeling and Multiple Mixtures. Mol Cell Proteomics. 2020;19(10):1706–23. Epub 2020/07/19. doi: 10.1074/mcp.RA120.002105 .
    1. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY, Geiger T, et al.. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13(9):731–40. Epub 2016/06/28. doi: 10.1038/nmeth.3901 .
    1. Seyfried NT, Dammer EB, Swarup V, Nandakumar D, Duong DM, Yin L, et al.. A Multi-network Approach Identifies Protein-Specific Co-expression in Asymptomatic and Symptomatic Alzheimer’s Disease. Cell Syst. 2017;4(1):60–72.e4. Epub 2016/12/15. doi: 10.1016/j.cels.2016.11.006 .
    1. Johnson ECB, Dammer EB, Duong DM, Ping L, Zhou M, Yin L, et al.. Large-scale proteomic analysis of Alzheimer’s disease brain and cerebrospinal fluid reveals early changes in energy metabolism associated with microglia and astrocyte activation. Nat Med. 2020;26(5):769–80. Epub 2020/04/13. doi: 10.1038/s41591-020-0815-6 .
    1. Simón-Manso Y, Lowenthal MS, Kilpatrick LE, Sampson ML, Telu KH, Rudnick PA, et al.. Metabolite profiling of a NIST Standard Reference Material for human plasma (SRM 1950): GC-MS, LC-MS, NMR, and clinical laboratory analyses, libraries, and web-based resources. Anal Chem. 2013;85(24):11725–31. Epub 20131203. doi: 10.1021/ac402503m .
    1. Smith CA, O’Maille G, Want EJ, Qin C, Trauger SA, Brandon TR, et al.. METLIN: a metabolite mass spectral database. Ther Drug Monit. 2005;27(6):747–51. doi: 10.1097/01.ftd.0000179845.53213.39 .
    1. Ludwig C, Gillet L, Rosenberger G, Amon S, Collins BC, Aebersold R. Data-independent acquisition-based SWATH-MS for quantitative proteomics: a tutorial. Mol Syst Biol. 2018;14(8):e8126. Epub 2018/08/15. doi: 10.15252/msb.20178126 .
    1. Bissler JJ, Kingswood JC, Radzikowska E, Zonnenberg BA, Belousova E, Frost MD, et al.. Everolimus long-term use in patients with tuberous sclerosis complex: Four-year update of the EXIST-2 study. PLoS One. 2017;12(8):e0180939. Epub 20170809. doi: 10.1371/journal.pone.0180939 .
    1. Pengel LH, Liu LQ, Morris PJ. Do wound complications or lymphoceles occur more often in solid organ transplant recipients on mTOR inhibitors? A systematic review of randomized controlled trials. Transpl Int. 2011;24(12):1216–30. Epub 20110929. doi: 10.1111/j.1432-2277.2011.01357.x .
    1. Roux PP, Shahbazian D, Vu H, Holz MK, Cohen MS, Taunton J, et al.. RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J Biol Chem. 2007;282(19):14056–64. Epub 20070314. doi: 10.1074/jbc.M700906200 .
    1. Mayer MP, Bukau B. Hsp70 chaperones: cellular functions and molecular mechanism. Cell Mol Life Sci. 2005;62(6):670–84. doi: 10.1007/s00018-004-4464-6 .
    1. Lackie RE, Maciejewski A, Ostapchenko VG, Marques-Lopes J, Choy WY, Duennwald ML, et al.. The Hsp70/Hsp90 Chaperone Machinery in Neurodegenerative Diseases. Front Neurosci. 2017;11:254. Epub 20170516. doi: 10.3389/fnins.2017.00254 .
    1. Yang T, Hsu C, Liao W, Chuang JS. Heat shock protein 70 expression in epilepsy suggests stress rather than protection. Acta Neuropathol. 2008;115(2):219–30. Epub 20071011. doi: 10.1007/s00401-007-0297-3 .
    1. Hu F, Zhou J, Lu Y, Guan L, Wei NN, Tang YQ, et al.. Inhibition of Hsp70 Suppresses Neuronal Hyperexcitability and Attenuates Epilepsy by Enhancing A-Type Potassium Current. Cell Rep. 2019;26(1):168–81.e4. doi: 10.1016/j.celrep.2018.12.032 .
    1. Thom M, Seetah S, Sisodiya S, Koepp M, Scaravilli F. Sudden and unexpected death in epilepsy (SUDEP): evidence of acute neuronal injury using HSP-70 and c-Jun immunohistochemistry. Neuropathol Appl Neurobiol. 2003;29(2):132–43. doi: 10.1046/j.1365-2990.2003.00452.x .
    1. Amanat S, Gallego-Martinez A, Sollini J, Perez-Carpena P, Espinosa-Sanchez JM, Aran I, et al.. Burden of rare variants in synaptic genes in patients with severe tinnitus: An exome based extreme phenotype study. EBioMedicine. 2021;66:103309. Epub 20210401. doi: 10.1016/j.ebiom.2021.103309 .
    1. Hata Y, Yoshida K, Kinoshita K, Nishida N. Epilepsy-related sudden unexpected death: targeted molecular analysis of inherited heart disease genes using next-generation DNA sequencing. Brain Pathol. 2017;27(3):292–304. Epub 2016/06/20. doi: 10.1111/bpa.12390 .
    1. Stankewich MC, Cianci CD, Stabach PR, Ji L, Nath A, Morrow JS. Cell organization, growth, and neural and cardiac development require αII-spectrin. J Cell Sci. 2011;124(Pt 23):3956–66. Epub 20111208. doi: 10.1242/jcs.080374 .
    1. Park SM, Lim JS, Ramakrishina S, Kim SH, Kim WK, Lee J, et al.. Brain Somatic Mutations in MTOR Disrupt Neuronal Ciliogenesis, Leading to Focal Cortical Dyslamination. Neuron. 2018;99(1):83–97.e7. Epub 20180621. doi: 10.1016/j.neuron.2018.05.039 .
    1. Takei N, Nawa H. mTOR signaling and its roles in normal and abnormal brain development. Front Mol Neurosci. 2014;7:28. Epub 20140423. doi: 10.3389/fnmol.2014.00028 .
    1. Brewster AL, Lugo JN, Patil VV, Lee WL, Qian Y, Vanegas F, et al.. Rapamycin reverses status epilepticus-induced memory deficits and dendritic damage. PLoS One. 2013;8(3):e57808. Epub 20130311. doi: 10.1371/journal.pone.0057808 .
    1. Morita M, Gravel SP, Chénard V, Sikström K, Zheng L, Alain T, et al.. mTORC1 controls mitochondrial activity and biogenesis through 4E-BP-dependent translational regulation. Cell Metab. 2013;18(5):698–711. doi: 10.1016/j.cmet.2013.10.001 .
    1. Hentze MW, Kulozik AE. A perfect message: RNA surveillance and nonsense-mediated decay. Cell. 1999;96(3):307–10. doi: 10.1016/s0092-8674(00)80542-5 .
    1. Martinez-Nunez RT, Wallace A, Coyne D, Jansson L, Rush M, Ennajdaoui H, et al.. Modulation of nonsense mediated decay by rapamycin. Nucleic Acids Res. 2017;45(6):3448–59. doi: 10.1093/nar/gkw1109 .
    1. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell. 2006;124(3):471–84. doi: 10.1016/j.cell.2006.01.016 .
    1. Figlia G, Gerber D, Suter U. Myelination and mTOR. Glia. 2018;66(4):693–707. Epub 20171206. doi: 10.1002/glia.23273 .
    1. Guo H, Zhong Y, Jackson AL, Clark LH, Kilgore J, Zhang L, et al.. Everolimus exhibits anti-tumorigenic activity in obesity-induced ovarian cancer. Oncotarget. 2016;7(15):20338–56. doi: 10.18632/oncotarget.7934 .
    1. Ramanathan A, Schreiber SL. Direct control of mitochondrial function by mTOR. Proc Natl Acad Sci U S A. 2009;106(52):22229–32. Epub 20091222. doi: 10.1073/pnas.0912074106 .
    1. Neale JH, Yamamoto T. N-acetylaspartylglutamate (NAAG) and glutamate carboxypeptidase II: An abundant peptide neurotransmitter-enzyme system with multiple clinical applications. Prog Neurobiol. 2020;184:101722. Epub 20191112. doi: 10.1016/j.pneurobio.2019.101722 .
    1. Morland C, Nordengen K. -Acetyl-Aspartyl-Glutamate in Brain Health and Disease. Int J Mol Sci. 2022;23(3). Epub 20220123. doi: 10.3390/ijms23031268 .
    1. Budde K, Zonnenberg BA, Frost M, Cheung W, Urva S, Brechenmacher T, et al.. Pharmacokinetics and pharmacodynamics of everolimus in patients with renal angiomyolipoma and tuberous sclerosis complex or lymphangioleiomyomatosis. Br J Clin Pharmacol. 2016;81(5):958–70. Epub 20160305. doi: 10.1111/bcp.12834 .
    1. Vitiello D, Neagoe PE, Sirois MG, White M. Effect of everolimus on the immunomodulation of the human neutrophil inflammatory response and activation. Cell Mol Immunol. 2015;12(1):40–52. Epub 20140602. doi: 10.1038/cmi.2014.24 .
    1. Martinet W, Verheye S, De Meyer I, Timmermans JP, Schrijvers DM, Van Brussel I, et al.. Everolimus triggers cytokine release by macrophages: rationale for stents eluting everolimus and a glucocorticoid. Arterioscler Thromb Vasc Biol. 2012;32(5):1228–35. Epub 20120216. doi: 10.1161/ATVBAHA.112.245381 .

Source: PubMed

3
구독하다