Мультиформная глиобластома (ГБМ) — наиболее агрессивная первичная опухоль головного мозга, характеризующаяся крайне неблагоприятным прогнозом. Трудности в диагностике и мониторинге данного заболевания создают необходимость поиска минимально инвазивных подходов, среди которых перспективным направлением считается жидкостная биопсия. Данный обзор посвящен анализу результатов современных исследований, направленных на поиск циркулирующих белковых биомаркеров ГБМ в плазме и сыворотке крови. В качестве биомаркеров рассматриваются свободно циркулирующие белки плазмы крови и белки, находящиеся в составе внеклеточных везикул (ВнВ). В обзоре обобщены результаты работ, использующих для поиска белковых биомаркеров как иммунохимические методы, так и масс-спектрометрические подходы, а также представлен перечень выявленных потенциальных диагностических и прогностических биомаркеров. Анализ представленных в литературе работ показывает, что протеомный анализ, сосредоточенный на фракции ВнВ плазмы крови, существенно расширяет возможности поиска биомаркеров для неинвазивной диагностики и мониторинга ГБМ.
Майоров В.А. и др. Циркулирующие белковые биомаркеры глиобластомы: методологические подходы и перспективы развития // Биомедицинская химия. - 2025. - Т. 71. -N 6. - С. .
Майоров В.А. и др., "Циркулирующие белковые биомаркеры глиобластомы: методологические подходы и перспективы развития." Биомедицинская химия 71.6 (2025): .
Майоров, В. А., Тихонова, О. В., Згода, В. Г. (2025). Циркулирующие белковые биомаркеры глиобластомы: методологические подходы и перспективы развития. Биомедицинская химия, 71(6), .
Список литературы
Grochans S., Cybulska A.M., Simińska D., Korbecki J., Kojder K., Chlubek D., Baranowska-Bosiacka I. (2022) Epidemiology of glioblastoma multiforme. Cancers (Basel), 14(10), 2412. CrossRef Scholar google search
Hanif F., Muzaffar K., Perveen K., Malhi S.M., Simjee S.U. (2017) Glioblastoma multiforme: a review of its epidemiology and pathogenesis through clinical presentation and treatment. Asian Pacific J. Cancer Prev., 18(1), 3–9. CrossRef Scholar google search
Tan A.C., Ashley D.M., López G.Y., Malinzak M., Friedman H.S., Khasraw M. (2020) Management of glioblastoma: state of the art and future directions. CA Cancer J. Clin., 70(4), 299–312. CrossRef Scholar google search
Ahmed R., Oborski M.J., Hwang M., Lieberman F.S., Mountz J.M. (2014) Malignant gliomas: current perspectives in diagnosis, treatment, and early response assessment using advanced quantitative imaging methods. Cancer Manag. Res., 24(6), 149–170. CrossRef Scholar google search
Ronvaux L., Riva M., Coosemans A., Herzog M., Rommelaere G., Donis N., d’Hondt L., Douxfils J. (2022) Liquid biopsy in glioblastoma. Cancers (Basel), 14(14), 3394. CrossRef Scholar google search
Gilard V., Tebani A., Dabaj I., Laquèrriere A., Fontanilles M., Derrey S., Marret S., Bekri S. (2021) Diagnosis and management of glioblastoma: a comprehensive perspective. J. Pers. Med., 11(4), 258. CrossRef Scholar google search
Seyhan A.A. (2024) Circulating liquid biopsy biomarkers in glioblastoma: advances and challenges. Int. J. Mol. Sci., 25(14), 7974. CrossRef Scholar google search
Eibl R.H., Schneemann M. (2023) Liquid biopsy and glioblastoma. Explor. Target Antitumor Ther., 4(1), 28–41. CrossRef Scholar google search
Jones J., Nguyen H., Drummond K., Morokoff A. (2021) Circulating biomarkers for glioma: a review. Neurosurgery, 88(3), E221–E230. CrossRef Scholar google search
Mo F., Pellerino A., Soffietti R., Rudà R. (2021) Blood-brain barrier in brain tumors: biology and clinical relevance. Int. J. Mol. Sci., 22(23), 12654. CrossRef Scholar google search
Constanzo F., Teixeira B.C.A., Sens P., Smaili H., Escuissato D.L., Ramina R. (2023) Perfusion-weighted imaging in vestibular schwannoma: the influence that cystic status and tumor size have on perfusion profiles. Radiol. Bras., 56(2), 67–74. CrossRef Scholar google search
Wu S.-K., Tsai C.-L., Huang Y., Hynynen K. (2021) Focused ultrasound and microbubbles-mediated drug delivery to brain tumor. Pharmaceutics, 13(1), 15. CrossRef Scholar google search
Arvanitis C.D., Ferraro G.B., Jain R.K. (2020) The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer, 20(1), 26–41. CrossRef Scholar google search
Upton D.H., Ung C., George S.M., Tsoli M., Kavallaris M., David S. (2022) Challenges and opportunities to penetrate the blood-brain barrier for brain cancer therapy. Theranostics, 12(10), 4734–4752. CrossRef Scholar google search
Digiovanni S., Lorenzati M., Bianciotto O.T., Godel M., Fontana S., Akman M., Costamagna C., Couraud P.-O., Buffo A., Kopecka J., Riganti C., Salaroglio I.C. (2024) Blood-brain barrier permeability increases with the differentiation of glioblastoma cells in vitro. Fluids Barriers CNS, 21, 89. CrossRef Scholar google search
Solier C., Langen H. (2014) Antibody-based proteomics and biomarker research — current status and limitations. Proteomics, 14(6), 774–783. CrossRef Scholar google search
Carlsson A., Persson O., Ingvarsson J., Widegren B., Salford L., Borrebaeck C.A.K., Wingren C. (2010) Plasma proteome profiling reveals biomarker patterns associated with prognosis and therapy selection in glioblastoma multiforme patients. Proteomics Clin. Appl., 4(6–7), 591–602. CrossRef Scholar google search
Ahir B.K., Engelhard H.H., Lakka S.S. (2020) Tumor development and angiogenesis in adult brain tumor: glioblastoma. Mol. Neurobiol., 57(5), 2461–2478. CrossRef Scholar google search
Xu C., Wu X., Zhu J. (2013) VEGF promotes proliferation of human glioblastoma multiforme stem-like cells through VEGF receptor 2. Sci. World J., 2013, 417413. CrossRef Scholar google search
Meier C., Brieger A. (2025) The role of IL-8 in cancer development and its impact on immunotherapy resistance. Eur. J. Cancer, 218, 115267. CrossRef Scholar google search
McClelland S., Maxwell P.J., Branco C., Barry S.T., Eberlein C., LaBonte M.J. (2024) Targeting IL-8 and its receptors in prostate cancer: inflammation, stress response, and treatment resistance. Cancers (Basel), 16(16), 2797. CrossRef Scholar google search
Urbantat R.M., Vajkoczy P., Brandenburg S. (2021) Advances in chemokine signaling pathways as therapeutic targets in glioblastoma. Cancers (Basel), 13(12), 2983. CrossRef Scholar google search
Elstner A., Stockhammer F., Nguyen-Dobinsky T.-N., Nguyen Q.L., Pilgermann I., Gill A., Guhr A., Zhang T., von Eckardstein K., Picht T., Veelken J., Martuza R.L., von Deimling A., Kurtz A. (2011) Identification of diagnostic serum protein profiles of glioblastoma patients. J. Neurooncol., 102(1), 71–80. CrossRef Scholar google search
Jung C.S., Foerch C., Schänzer A., Heck A., Plate K.H., Seifert V., Steinmetz H., Raabe A., Sitzer M. (2007) Serum GFAP is a diagnostic marker for glioblastoma multiforme. Brain, 130(12), 3336–3341. CrossRef Scholar google search
Tichy J., Spechtmeyer S., Mittelbronn M., Hattingen E., Rieger J., Senft C., Foerch C. (2015) Prospective evaluation of serum glial fibrillary acidic protein (GFAP) as a diagnostic marker for glioblastoma. J. Neurooncol., 126(2), 361–369. CrossRef Scholar google search
Kiviniemi A., Gardberg M., Frantzén J., Parkkola R., Vuorinen V., Pesola M., Minn H. (2015) Serum levels of GFAP and EGFR in primary and recurrent high-grade gliomas: correlation to tumor volume, molecular markers, and progression-free survival. J. Neurooncol., 124(2), 237–245. CrossRef Scholar google search
Iwamoto F.M., Hottinger A.F., Karimi S., Riedel E., Dantis J., Jahdi M., Panageas K.S., Lassman A.B., Abrey L.E., Fleisher M., DeAngelis L.M., Holland E.C., Hormigo A. (2011) Serum YKL-40 is a marker of prognosis and disease status in high-grade gliomas. Neuro. Oncol., 13(11), 1244–1251. CrossRef Scholar google search
Bernardi D., Padoan A., Ballin A., Sartori M.T., Manara R., Scienza R., Plebani M., Della Puppa A. (2012) Serum YKL-40 following resection for cerebral glioblastoma. J. Neurooncol., 107(2), 299–305. CrossRef Scholar google search
Shao R. (2013) YKL-40 acts as an angiogenic factor to promote tumor angiogenesis. Front. Physiol., 4(122), 122. CrossRef Scholar google search
Francescone R.A., Scully S., Faibish M., Taylor S.L., Oh D., Moral L., Yan W., Bentley B., Shao R. (2011) Role of YKL-40 in the angiogenesis, radioresistance, and progression of glioblastoma. J. Biol. Chem., 286(17), 15332–15343. CrossRef Scholar google search
Francescone R., Ngernyuang N., Yan W., Bentley B., Shao R. (2014) Tumor-derived mural-like cells coordinate with endothelial cells: role of YKL-40 in mural cell-mediated angiogenesis. Oncogene, 33(16), 2110–2122. CrossRef Scholar google search
Boisen M.K., Holst C.B., Consalvo N., Chinot O.L., Johansen J.S. (2018) Plasma YKL-40 as a biomarker for bevacizumab efficacy in patients with newly diagnosed glioblastoma in the phase 3 randomized AVAglio trial. Oncotarget, 9(6), 6752–6762. CrossRef Scholar google search
Shao R., Francescone R., Ngernyuang N., Bentley B., Taylor S.L., Moral L., Yan W. (2014) Anti-YKL-40 antibody and ionizing irradiation synergistically inhibit tumor vascularization and malignancy in glioblastoma. Carcinogenesis, 35(2), 373–382. CrossRef Scholar google search
Lin Y., Wang J.F., Gao G.Z., Zhang G.Z., Wang F.L., Wang Y.J. (2013) Plasma levels of tissue inhibitor of matrix metalloproteinase-1 correlate with diagnosis and prognosis of glioma patients. Chin. Med. J., 126(22), 4295–4300. CrossRef Scholar google search
Ramachandran R.K., Swrensen M.D., Aaberg-Jessen C., Hermansen S.K., Kristensen B.W. (2017) Expression and prognostic impact of matrix metalloproteinase-2 (MMP-2) in astrocytomas. PLOS One, 12(2), e0172234. CrossRef Scholar google search
Aaberg-Jessen C., Halle B., Jensen S.S., Müller S., Rømer U.M., Pedersen C.B., Brünner N., Kristensen B.W. (2016) Comparative studies of TIMP-1 immunohistochemistry, TIMP-1 FISH analysis and plasma TIMP-1 in glioblastoma patients. J. Neurooncol., 130(3), 439–448. CrossRef Scholar google search
Taheri E., Raeeszadeh-Sarmazdeh M. (2025) Effect of TIMPs and their minimally engineered variants in blocking invasion and migration of brain cancer cells. Oncotarget, 16, 118–130. CrossRef Scholar google search
Zupancic K., Blejec A., Herman A., Veber M., Verbovsek U., Korsic M., Knezevic M., Rozman P., Turnsek T.L., Gruden K., Motaln H. (2014) Identification of plasma biomarker candidates in glioblastoma using an antibody-array-based proteomic approach. Radiol. Oncol., 48(3), 257–266. CrossRef Scholar google search
Du M., Feng J., Tao Y., Pan Q., Chen F. (2021) GNAO1 as a novel predictive biomarker for late relapse in hepatocellular carcinoma. J. Healthc. Eng., 2021, 7631815. CrossRef Scholar google search
Sun B., Wang G., Chen G., Zhang Y., Yang R., Hua H., Li Y., Feng H. (2025) GNAO1 overexpression promotes neural differentiation of glioma stem-like cells and reduces tumorigenicity through TRIM21/CREB/HES1 axis. Oncogene, 44(7), 450–461. CrossRef Scholar google search
Geyer P.E., Holdt L.M., Teupser D., Mann M. (2017) Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol., 13(9), 942. CrossRef Scholar google search
Rehiman S.H., Lim S.M., Neoh C.F., Majeed A.B.A., Chin A.-V., Tan M.P., Kamaruzzaman S.B., Ramasamy K. (2020) Proteomics as a reliable approach for discovery of blood-based Alzheimer’s disease biomarkers: a systematic review and meta-analysis. Ageing Res. Rev., 60, 101066. CrossRef Scholar google search
Gollapalli K., Ray S., Srivastava R., Renu D., Singh P., Dhali S., Bajpai Dikshit J., Srikanth R., Moiyadi A., Srivastava S. (2012) Investigation of serum proteome alterations in human glioblastoma multiforme. Proteomics, 12(14), 2378–2390. CrossRef Scholar google search
Miyauchi E., Furuta T., Ohtsuki S., Tachikawa M., Uchida Y., Sabit H., Obuchi W., Baba T., Watanabe M., Terasaki T., Nakada M. (2018) Identification of blood biomarkers in glioblastoma by SWATH mass spectrometry and quantitative targeted absolute proteomics. PLOS One, 13(3), e0193799. CrossRef Scholar google search
Furuta T., Sugita Y., Komaki S., Ohshima K., Morioka M., Uchida Y., Tachikawa M., Ohtsuki S., Terasaki T., Nakada M. (2020) The multipotential of leucine-rich α-2 glycoprotein 1 as a clinicopathological biomarker of glioblastoma. J. Neuropathol. Exp. Neurol., 79(8), 873–879. CrossRef Scholar google search
Furuta T., Miyoshi H., Moritsubo M., Nakajima R., Negoto T., Nakamura H., Morioka M., Uchida Y., Ohtsuki S., Nakada M. (2025) Biological role and clinicopathological significance of leucine-rich α-2 glycoprotein 1 in the glioblastoma microenvironment. J. Neuropathol. Exp. Neurol., 84(8), 707–714. CrossRef Scholar google search
Kumar D.M., Thota B., Shinde S.V., Prasanna K.V., Hegde A.S., Arivazhagan A., Chandramouli B.A., Santosh V., Somasundaram K. (2010) Proteomic identification of haptoglobin α2 as a glioblastoma serum biomarker: implications in cancer cell migration and tumor growth. J. Proteome Res., 9(11), 5557–5567. CrossRef Scholar google search
Naryzhny S., Ronzhina N., Zorina E., Kabachenko F., Zavialova M., Zgoda V., Klopov N., Legina O., Pantina R. (2021) Evaluation of haptoglobin and its proteoforms as glioblastoma markers. Int. J. Mol. Sci., 22(12), 6533. CrossRef Scholar google search
Odiase P., Ma J., Ranganathan S., Ogunkua O., Turner W.B., Marshall D., Ochieng J. (2024) The role of fetuin-A in tumor cell growth, prognosis, and dissemination. Int. J. Mol. Sci., 25(23), 12918. CrossRef Scholar google search
Petrik V., Saadoun S., Loosemore A., Hobbs J., Opstad K.S., Sheldon J., Tarelli E., Howe F.A., Bell B.A., Papadopoulos M.C. (2008) Serum α2-HS glycoprotein predicts survival in patients with glioblastoma. Clin. Chem., 54(4), 713–722. CrossRef Scholar google search
van Linde M.E., van der Mijn J.C., Pham T.V., Knol J.C., Wedekind L.E., Hovinga K.E., Aliaga E.S., Buter J., Jimenez C.R., Reijneveld J.C., Verheul H.M.W. (2016) Evaluation of potential circulating biomarkers for prediction of response to chemoradiation in patients with glioblastoma. J. Neurooncol., 129(2), 221–230. CrossRef Scholar google search
Ponomarenko E.A., Poverennaya E.V., Ilgisonis E.V., Pyatnitskiy M.A., Kopylov A.T., Zgoda V.G., Lisitsa A.V., Archakov A.I. (2016) The size of the human proteome: the width and depth. Int. J. Anal. Chem., 2016, 7436849. CrossRef Scholar google search
Gautam P., Nair S.C., Gupta M.K., Sharma R., Polisetty R.V., Chandak G.R., Harsha H.C., Sirdeshmukh R. (2012) Proteins with altered levels in plasma from glioblastoma patients as revealed by iTRAQ-based quantitative proteomic analysis. PLOS One, 7(9), e46153. CrossRef Scholar google search
Ji Q., Li Z., Guo Y., Zhang X. (2024) S100A9, as a potential predictor of prognosis and immunotherapy response for GBM, promotes the malignant progression of GBM cells and migration of M2 macrophages. Aging (Albany N.Y.), 16(15), 11513–11534. CrossRef Scholar google search
Kalluri R. (2016) The biology and function of exosomes in cancer. J. Clin. Invest., 126(4), 1208–1215. CrossRef Scholar google search
El Andaloussi S., Mäger I., Breakefield X.O., Wood M.J.A. (2013) Extracellular vesicles: biology and emerging therapeutic opportunities. Nat. Rev. Drug Discov., 12(5), 347–357. CrossRef Scholar google search
Kalluri R. (2024) The biology and function of extracellular vesicles in immune response and immunity. Immunity, 57(8), 1752–1768. CrossRef Scholar google search
Chiaradia E., Tancini B., Emiliani C., Delo F., Pellegrino R.M., Tognoloni A., Urbanelli L., Buratta S. (2021) Extracellular vesicles under oxidative stress conditions: biological properties and physiological roles. Cells, 10(7), 1763. CrossRef Scholar google search
Maacha S., Bhat A.A., Jimenez L., Raza A., Haris M., Uddin S., Grivel J.-C. (2019) Extracellular vesiclesmediated intercellular communication: roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer, 18, 55. CrossRef Scholar google search
Ortiz A. (2021) Extracellular vesicles in cancer progression. Semin. Cancer Biol., 76(3), 139–142. CrossRef Scholar google search
Marar C., Starich B., Wirtz D. (2021) Extracellular vesicles in immunomodulation and tumor progression. Nat. Immunol., 22(5), 560–570. CrossRef Scholar google search
Zhang C., Qin C., Dewanjee S., Bhattacharya H., Chakraborty P., Jha N.K., Gangopadhyay M., Jha S.K., Liu Q. (2024) Tumor-derived small extracellular vesicles in cancer invasion and metastasis: molecular mechanisms, and clinical significance. Mol. Cancer, 23, 18. CrossRef Scholar google search
Ramos-Zaldívar H.M., Polakovicova I., Salas-Huenuleo E., Corvalán A.H., Kogan M.J., Yefi C.P., Andia M.E. (2022) Extracellular vesicles through the blood-brain barrier: a review. Fluids Barriers CNS, 19, 60. CrossRef Scholar google search
Saint-Pol J., Gosselet F., Duban-Deweer S., Pottiez G., Karamanos Y. (2020) Targeting and crossing the blood-brain barrier with extracellular vesicles. Cells, 9(4), 851. CrossRef Scholar google search
Kalluri R., McAndrews K.M. (2023) The role of extracellular vesicles in cancer. Cell, 186(8), 1610–1626. CrossRef Scholar google search
Li J., He X., Deng Y., Yang C. (2019) An update on isolation methods for proteomic studies of extracellular vesicles in biofluids. Molecules, 24(19), 3516. CrossRef Scholar google search
Sidhom K., Obi P.O., Saleem A. (2020) A review of exosomal isolation methods: is size exclusion chromatography the best option? Int. J. Mol. Sci., 21(18), 6466. CrossRef Scholar google search
Veerman R.E., Teeuwen L., Czarnewski P., Güclüler Akpinar G., Sandberg A.S., Cao X., Pernemalm M., Orre L.M., Gabrielsson S., Eldh M. (2021) Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J. Extracell. Vesicles, 10(9), e12128. CrossRef Scholar google search
Xiao J. (2023) Updated methods of extracellular vesicles isolation. Adv. Exp. Med. Biol., 1418, 3–14. CrossRef Scholar google search
Suresh P.S., Zhang Q. (2025) Comprehensive comparison of methods for isolation of extracellular vesicles from human plasma. J. Proteome Res., 24(6), 2956–2967. CrossRef Scholar google search
Welsh J.A., Goberdhan D.C.I., O’Driscoll L., Buzas E.I., Blenkiron C., Bussolati B., Cai H., di Vizio D., Driedonks T.A.P., Erdbrügger U., Falcon-Perez J.M., Fu Q.L., Hill A.F., Lenassi M., Lim S.K., Mahoney M.G., Mohanty S., Möller A., Nieuwland R., Ochiya T., Sahoo S., Torrecilhas A.C., Zheng L., Zijlstra A., Abuelreich S., Bagabas R., Bergese P., Bridges E.M., Brucale M., Burger D., Carney R.P., Cocucci E., Crescitelli R., Hanser E., Harris A.L., Haughey N.J., Hendrix A., Ivanov A.R., Jovanovic-Talisman T., Kruh-Garcia N.A., Ku'ulei-Lyn Faustino V., Kyburz D., Lässer C., Lennon K.M., Lötvall J., Maddox A.L., Martens-Uzunova E.S., Mizenko R.R., Newman L.A., Ridolfi A., Rohde E., Rojalin T., Rowland A., Saftics A., Sandau U.S., Saugstad J.A., Shekari F., Swift S., Ter-Ovanesyan D., Tosar J.P., Useckaite Z., Valle F., Varga Z., van der Pol E., van Herwijnen M.J.C., Wauben M.H.M., Wehman A.M., Williams S., Zendrini A., Zimmerman A.J.; MISEV Consortium; Théry C., Witwer K.W. (2024) Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J. Extracell. Vesicles, 13(2), e12404. CrossRef Scholar google search
Lane R., Simon T., Vintu M., Solkin B., Koch B., Stewart N., Benstead-Hume G., Pearl F.M.G., Critchley G., Stebbing J., Giamas G. (2019) Cell-derived extracellular vesicles can be used as a biomarker reservoir for glioblastoma tumor subtyping. Commun. Biol., 2, 315. CrossRef Scholar google search
Binda E., Visioli A., Giani F., Trivieri N., Palumbo O., Restelli S., Dezi F., Mazza T., Fusilli C., Legnani F., Carella M., di Meco F., Duggal R., Vescovi A.L. (2017) Wnt5a drives an invasive phenotype in human glioblastoma stem-like cells. Cancer Res., 77(4), 996–1007. CrossRef Scholar google search
Guo S.-K., Shen M.-F., Yao H.-W., Liu Y.-S. (2018) Enhanced expression of TGFBI promotes the proliferation and migration of glioma cells. Cell. Physiol. Biochem., 49(3), 1097–1109. CrossRef Scholar google search
Peng P., Zhu H., Liu D., Chen Z., Zhang X., Guo Z., Dong M., Wan L., Zhang P., Liu G., Zhang S., Dong F., Hu F., Cheng F., Huang S., Guo D., Zhang B., Yu X., Wan F. (2022) TGFBI secreted by tumor-associated macrophages promotes glioblastoma stem cell-driven tumor growth via integrin αvβ5-Src-Stat3 signaling. Theranostics, 12(9), 4221–4236. CrossRef Scholar google search
Zhang Y., Xiao X., Yang G., Jiang X., Jiao S., Nie Y., Zhang T. (2025) STAT3/TGFBI signaling promotes the temozolomide resistance of glioblastoma through upregulating glycolysis by inducing cellular senescence. Cancer Cell Int., 25, 127. CrossRef Scholar google search
Chédeville A.L., Lourdusamy A., Monteiro A.R., Hill R., Madureira P.A. (2020) Investigating glioblastoma response to hypoxia. Biomedicines, 8(9), 310. CrossRef Scholar google search
Codó P., Weller M., Kaulich K., Schraivogel D., Silginer M., Reifenberger G., Meister G., Roth P. (2016) Control of glioma cell migration and invasiveness by GDF-15. Oncotarget, 7(7), 7732–7746. CrossRef Scholar google search
Bentaberry-Rosa A., Nicaise Y., Delmas C., Gouazé-Andersson V., Cohen-Jonathan-Moyal E., Seva C. (2024) Overexpression of growth differentiation factor 15 in glioblastoma stem cells promotes their radioresistance. Cancers (Basel), 16(1), 27. CrossRef Scholar google search
Naryzhny S., Volnitskiy A., Kopylov A., Zorina E., Kamyshinsky R., Bairamukov V., Garaeva L., Shlikht A., Shtam T. (2020) Proteome of glioblastoma-derived exosomes as a source of biomarkers. Biomedicines, 8(7), 216. CrossRef Scholar google search
Нарыжный С.Н., Ронжина Н.Л., Майнскова М.А., Белякова Н.В., Пантина Р.А., Филатов М.В. (2014) Разработка штрих-кода и получение белкового профиля глиобластомы. Биомедицинская химия, 60(3), 308–321. CrossRef Scholar google search
Anastasi F., Greco F., Dilillo M., Vannini E., Cappello V., Baroncelli L., Costa M., Gemmi M., Caleo M., McDonnell L.A. (2020) Proteomics analysis of serum small extracellular vesicles for the longitudinal study of a glioblastoma multiforme mouse model. Sci. Rep., 10, 20498. CrossRef Scholar google search
Greco F., Anastasi F., Pardini L.F., Dilillo M., Vannini E., Baroncelli L., Caleo M., McDonnell L.A. (2021) Longitudinal bottom-up proteomics of serum, serum extracellular vesicles, and cerebrospinal fluid reveals candidate biomarkers for early detection of glioblastoma in a murine model. Molecules, 26(19), 5992. CrossRef Scholar google search
Indira Chandran V., Welinder C., Mansson A.S., Offer S., Freyhult E., Pernemalm M., Lund S.M., Pedersen S., Lehtio J., Marko-Varga G., Johansson M.C., Englund E., Sundgren P.C., Belting M. (2019) Ultrasensitive immunoprofiling of plasma extracellular vesicles identifies syndecan-1 as a potential tool for minimally invasive diagnosis of glioma. Clin. Cancer Res., 25(10), 3115–3127. CrossRef Scholar google search
Luce A., Abate M., Scognamiglio G., Montella M., Iervolino D., Campione S., di Mauro A., Sepe O., Gigantino V., Tathode M.S., Ferrara G., Monaco R., de Dominicis G., Misso G., Gentile V., Franco R., Zappavigna S., Caraglia M. (2024) Immune cell infiltration and inflammatory landscape in primary brain tumours. J. Transl. Med., 22, 521. CrossRef Scholar google search
Zheng W., Chen Q., Liu H., Zeng L., Zhou Y., Liu X., Bai Y., Zhang J., Pan Y., Shao C. (2023) SDC1-dependent TGM2 determines radiosensitivity in glioblastoma by coordinating EPG5-mediated fusion of autophagosomes with lysosomes. Autophagy, 19(3), 839–857. CrossRef Scholar google search
Zeng L., Zheng W., Liu X., Zhou Y., Jin X., Xiao Y., Bai Y., Pan Y., Zhang J., Shao C. (2023) SDC1-TGM2-FLOT1-BHMT complex determines radio sensitivity of glioblastoma by influencing the fusion of autophagosomes with lysosomes. Theranostics, 13(11), 3725–3743. CrossRef Scholar google search
Osti D., del Bene M., Rappa G., Santos M., Matafora V., Richichi C., Faletti S., Beznoussenko G.V., Mironov A., Bachi A., Fornasari L., Bongetta D., Gaetani P., DiMeco F., Lorico A., Pelicci G. (2019) Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin. Cancer Res., 25(1), 266–276. CrossRef Scholar google search
Zhu H., Yu X., Zhang S., Shu K. (2021) Targeting the complement pathway in malignant glioma microenvironments. Front. Cell Dev. Biol., 9, 657472. CrossRef Scholar google search
Hallal S., Azimi A., Wei H., Ho N., Lee M.Y.T., Sim H.-W., Sy J., Shivalingam B., Buckland M.E., Alexander-Kaufman K.L. (2020) A comprehensive proteomic SWATH-MS workflow for profiling blood extracellular vesicles: a new avenue for glioma tumour surveillance. Int. J. Mol. Sci., 21(13), 4754. CrossRef Scholar google search
Mallawaaratchy D.M., Hallal S., Russell B., Ly L., Ebrahimkhani S., Wei H., Christopherson R.I., Buckland M.E., Kaufman K.L. (2017) Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease. J. Neurooncol., 131(2), 233–244. CrossRef Scholar google search
Liu G., Zhang P., Chen S., Chen Z., Qiu Y., Peng P., Huang W., Cheng F., Zhang Y., Li H., Xiao Q., Mao F., Wang B., Jiang X., Wan F., Guo D., Yu X. (2023) FAM129A promotes self-renewal and maintains invasive status via stabilizing the Notch intracellular domain in glioma stem cells. Neuro. Oncol., 25(10), 1788–1801. CrossRef Scholar google search
Шушкова Н.А., Вавилов Н.Э., Новикова С.Е., Фарафонова Т.Е., Тихонова О.В., Лияо Пао-чи, Згода В.Г. (2018) Количественный протеомный анализ экзосом крови человека. Биомедицинская химия, 64(6), 496–504. CrossRef Scholar google search
Novikova S.E., Soloveva N.A., Farafonova T.E., Tikhonova O.V., Liao P.-C., Zgoda V.G. (2021) Proteomic signature of extracellular vesicles for lung cancer recognition. Molecules, 26(20), 6145. CrossRef Scholar google search
Soloveva N., Novikova S., Farafonova T., Tikhonova O., Zgoda V. (2023) Proteomic signature of extracellular vesicles associated with colorectal cancer. Molecules, 28(10), 4227. CrossRef Scholar google search
Soloveva N., Novikova S., Farafonova T., Tikhonova O., Zgoda V. (2025) Secretome and proteome of extracellular vesicles provide protein markers of lung and colorectal cancer. Int. J. Mol. Sci., 26(3), 1016. CrossRef Scholar google search