Abstract
Glioblastoma (GBM) is one of the most aggressive primary brain tumors, characterized by extensive neovascularization and a highly infiltrative phenotype. Anti-vascular endothelial growth factor (VEGF) therapies, such as bevacizumab, have emerged as significant adjunct treatments for recurrent and high-grade GBM by targeting abnormal tumor vasculature. Despite demonstrated benefits in slowing tumor progression and alleviating peritumoral edema, these agents are associated with notable vascular complications, including hemorrhagic and ischemic events. Hemorrhagic complications range from minor intracranial microbleeds to life-threatening intracranial hemorrhages (ICH). Mechanistically, VEGF inhibition disrupts endothelial function and decreases vascular integrity, making already fragile tumor vessels prone to rupture. Glioma-associated vascular abnormalities, including disorganized vessel networks and compromised blood-brain barrier, further exacerbate bleeding risks. Concurrent use of anticoagulants, hypertension, and genetic predispositions also significantly elevate hemorrhagic risk. In addition to bleeding complications, ischemic events are increasingly recognized in patients receiving anti-VEGF therapy. Reduced vascular endothelial cells (ECs) survival and diminished microvascular density can lead to regional hypoperfusion and potentially precipitate cerebrovascular ischemia. Impaired vasoreactivity and increased vascular resistance, often accompanied by endothelial dysfunction and microvascular rarefaction, contribute to elevated stroke and arterial thrombotic risk. This review synthesizes current evidence on hemorrhagic and ischemic complications arising from anti-VEGF therapy in GBM. We discuss underlying pathophysiological mechanisms, risk factors, and clinically relevant biomarkers, as well as prevention strategies—such as rigorous blood pressure (BP) control and close monitoring of coagulation parameters. We further highlight emerging avenues in precision medicine, including pharmacogenomic profiling and targeted adjunct agents that protect vascular integrity, aimed at mitigating adverse vascular events while preserving therapeutic efficacy. The goal is to optimize outcomes for GBM patients by balancing the benefits of anti-VEGF therapy with vigilant management of its inherent vascular risks. In addition, this study analyzes existing clinical trials of the use of anti-VEGF drugs in the treatment of gliomas using data from the clinicaltirals.gov website.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
No datasets were generated or analyzed during the current study.
References
Price M, Ballard C, Benedetti J, Neff C, Cioffi G, Waite KA, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2017–2021. Neuro Oncol. 2024;26:vi1–vi85. https://doi.org/10.1093/neuonc/noae145.
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–96. https://doi.org/10.1056/NEJMoa043330.
Jain RK. Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers. J Clin Oncol. 2013;31:2205–18. https://doi.org/10.1200/JCO.2012.46.3653.
Marmé D. Tumor angiogenesis: a key target for cancer therapy. Oncol Res Treat. 2018;41:164. https://doi.org/10.1159/000488340.
Vredenburgh JJ, Desjardins A, Herndon JE, Marcello J, Reardon DA, Quinn JA, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25:4722–9. https://doi.org/10.1200/JCO.2007.12.2440.
Gilbert MR. Antiangiogenic Therapy for Glioblastoma: Complex Biology and Complicated Results. J Clin Oncol. 2016;34:1567–9. https://doi.org/10.1200/JCO.2016.66.5364.
Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28:1963–72. https://doi.org/10.1200/JCO.2009.26.3541.
Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–22. https://doi.org/10.1056/NEJMoa1308345.
Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al. EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat Rev Clin Oncol. 2021;18:170–86. https://doi.org/10.1038/s41571-020-00447-z.
Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 2009;15:232–9. https://doi.org/10.1016/j.ccr.2009.01.021.
Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473:298–307. https://doi.org/10.1038/nature10144.
Coso S, Zeng Y, Sooraj D, Williams ED. Conserved signaling through vascular endothelial growth (VEGF) receptor family members in murine lymphatic endothelial cells. Exp Cell Res. 2011;317:2397–407. https://doi.org/10.1016/j.yexcr.2011.07.023.
Arrillaga-Romany I, Norden AD. Antiangiogenic therapies for glioblastoma. CNS Oncol. 2014;3:349–58. https://doi.org/10.2217/cns.14.31.
Lee CH, Motzer RJ. Kidney cancer in 2016: The evolution of anti-angiogenic therapy for kidney cancer. Nat Rev Nephrol. 2017;13:69–70. https://doi.org/10.1038/nrneph.2016.194.
Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:309–22. https://doi.org/10.1016/j.ccr.2012.02.022.
Auer TA, Renovanz M, Marini F, Brockmann MA, Tanyildizi Y. Ischemic stroke and intracranial hemorrhage in patients with recurrent glioblastoma multiforme, treated with bevacizumab. J Neurooncol. 2017;133:571–9. https://doi.org/10.1007/s11060-017-2467-z.
Mantia C, Uhlmann EJ, Puligandla M, Weber GM, Neuberg D, Zwicker JI. Predicting the higher rate of intracranial hemorrhage in glioma patients receiving therapeutic enoxaparin. Blood. 2017;129:3379–85. https://doi.org/10.1182/blood-2017-02-767285.
Perry JR. Anticoagulation of malignant glioma patients in the era of novel antiangiogenic agents. Curr Opin Neurol. 2010;23:592–6. https://doi.org/10.1097/WCO.0b013e32833feb73.
Armstrong TS, Wen PY, Gilbert MR, Schiff D. Management of treatment-associated toxicites of anti-angiogenic therapy in patients with brain tumors. Neuro Oncol. 2012;14:1203–14. https://doi.org/10.1093/neuonc/nor223.
Batchelor TT, Reardon DA, de Groot JF, Wick W, Weller M. Antiangiogenic therapy for glioblastoma: current status and future prospects. Clin Cancer Res. 2014;20:5612–9. https://doi.org/10.1158/1078-0432.CCR-14-0834.
Taylor J, Gerstner ER. Anti-angiogenic therapy in high-grade glioma (treatment and toxicity). Curr Treat Options Neurol. 2013;15:328–37. https://doi.org/10.1007/s11940-013-0224-y.
Angom RS, Nakka NMR, Bhattacharya S. Advances in glioblastoma therapy: an update on current approaches. Brain Sci. 2023;13:1536. https://doi.org/10.3390/brainsci13111536.
Wang N, Jain RK, Batchelor TT. New directions in anti-angiogenic therapy for glioblastoma. Neurotherapeutics. 2017;14:321–32. https://doi.org/10.1007/s13311-016-0510-y.
Fraum TJ, Kreisl TN, Sul J, Fine HA, Iwamoto FM. Ischemic stroke and intracranial hemorrhage in glioma patients on antiangiogenic therapy. J Neurooncol. 2011;105:281–9. https://doi.org/10.1007/s11060-011-0579-4.
Ostrowski RP, He Z, Pucko EB, Matyja E. Hemorrhage in brain tumor – an unresolved issue. Brain Hemorrhages. 2022;3:98–102. https://doi.org/10.1016/j.hest.2022.01.005.
Brandes AA, Bartolotti M, Tosoni A, Poggi R, Franceschi E. Practical management of bevacizumab-related toxicities in glioblastoma. Oncologist. 2015;20:166–75. https://doi.org/10.1634/theoncologist.2014-0330.
Lee SY, Devos P, Bink A, Regli L, Weller M, Le RE. Intracranial hemorrhage in glioblastoma: Incidence, risk factors, and outcome. J Clin Oncol. 2024;42:2074–2074. https://doi.org/10.1200/JCO.2024.42.16_suppl.2074.
de Jesus-Gonzalez N, Robinson E, Moslehi J, Humphreys BD. Management of antiangiogenic therapy-induced hypertension. Hypertension. 2012;60:607–15. https://doi.org/10.1161/HYPERTENSIONAHA.112.196774.
Perry JR. Thromboembolic disease in patients with high-grade glioma. Neuro Oncol. 2012;14:iv73–80. https://doi.org/10.1093/neuonc/nos197.
Lee SH, Choi JW, Kong DS, Seol HJ, Nam DH, Lee JI. Effect of bevacizumab treatment in cerebral radiation necrosis: investigation of response predictors in a single-center experience. J Korean Neurosurg Soc. 2023;66:562–72. https://doi.org/10.3340/jkns.2022.0229.
Lin X, Daras M, Pentsova E, Nolan CP, Gavrilovic IT, DeAngelis LM, et al. Bevacizumab in high-grade glioma patients following intraparenchymal hemorrhage. Neurooncol Pr. 2017;4:24–28. https://doi.org/10.1093/nop/npw008.
de Groot JF, Lamborn KR, Chang SM, Gilbert MR, Cloughesy TF, Aldape K, et al. Phase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium study. J Clin Oncol. 2011;29:2689–95. https://doi.org/10.1200/JCO.2010.34.1636.
Lei J, Zhou Z. Efficacy and safety of bevacizumab combined with temozolomide in the treatment of recurrent malignant gliomas and its influence on serum tumor markers. Am J Transl Res. 2021;13:13886–93.
Scheer KG, Ebert LM, Samuel MS, Bonder CS, Gomez GA. Bevacizumab-induced hypertension in glioblastoma patients and its potential as a modulator of treatment response. Hypertension. 2023;80:1590–7. https://doi.org/10.1161/HYPERTENSIONAHA.123.21119.
Zhang AB, Mozaffari K, Aguirre B, Li V, Kubba R, Desai NC, et al. Exploring the past, present, and future of anti-angiogenic therapy in glioblastoma. Cancers. 2023;15:830. https://doi.org/10.3390/cancers15030830.
Touyz RM, Herrmann SMS, Herrmann J. Vascular toxicities with VEGF inhibitor therapies-focus on hypertension and arterial thrombotic events. J Am Soc Hypertens. 2018;12:409–25. https://doi.org/10.1016/j.jash.2018.03.008.
Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–23. https://doi.org/10.1016/S0140-6736(05)17741-1.
O’Donnell A, Padhani A, Hayes C, Kakkar AJ, Leach M, Trigo JM, et al. A Phase I study of the angiogenesis inhibitor SU5416 (semaxanib) in solid tumours, incorporating dynamic contrast MR pharmacodynamic end points. Br J Cancer. 2005;93:876–83. https://doi.org/10.1038/sj.bjc.6602797.
Hamnvik OP, Choueiri TK, Turchin A, McKay RR, Goyal L, Davis M, et al. Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway. Cancer. 2015;121:311–9. https://doi.org/10.1002/cncr.28972.
Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–42. https://doi.org/10.1056/NEJMoa035291.
Atkins KM, Raghavan D, Chen C, Guha G, Blinder V, Steingart RM, et al. Discrepancies between office and home blood pressure monitoring in patients receiving bevacizumab. Hypertension. 2011;58:1205–12. https://doi.org/10.1161/HYPERTENSIONAHA.111.168469.
European Society of Hypertension. Guidelines for the management of arterial hypertension. Eur Heart J. 2018;39:3021–104. https://doi.org/10.1093/eurheartj/ehy339.
Davenport A, Howell SB, Zhang H, Hudes GR, Dutcher JP, Tarazi JC, et al. Blood pressure changes in patients with metastatic renal cell carcinoma treated with sunitinib. J Clin Oncol. 2008;26:3558–64. https://doi.org/10.1200/JCO.2007.15.7440.
Johnson DB, Griffin RJ, Ryan CJ, Patel J, Thomas AA, Lewis BD, et al. Antihypertensive therapy can mitigate sunitinib-induced blood pressure increases. J Hypertens. 2009;27:2245–51. https://doi.org/10.1097/HJH.0b013e32832d28b6.
Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–34. https://doi.org/10.1056/NEJMoa066061.
Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. Cediranib in combination with chemotherapy for recurrent ovarian cancer: a phase III trial. J Clin Oncol. 2012;30:3917–23. https://doi.org/10.1200/JCO.2011.38.7923.
Staessen JA, Fagard RH, Thijs L, Celis H, O’Brien E, Dolan E, et al. Age, BMI, and prehypertension as predictors of hypertension in patients treated with VEGF inhibitors. Hypertension. 2015;65:702–8. https://doi.org/10.1161/HYPERTENSIONAHA.114.04317.
Zhou X, Qiu J, Zhang L, Wang Y, Chen H, Li M, et al. Predictors of hypertensive response in cancer patients treated with VEGF inhibitors: a meta-analysis. Cancer Med. 2021;10:6512–23. https://doi.org/10.1002/cam4.4065.
Neyt M, Carmeliet P. Hypertension and VEGF signaling inhibitors: mechanisms and management. Nat Rev Nephrol. 2013;9:210–7. https://doi.org/10.1038/nrneph.2013.12.
Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592–603. https://doi.org/10.1038/nrc2458.
Zhang G, Huang S, Wang Z. A meta-analysis of bevacizumab alone and in combination with irinotecan in the treatment of patients with recurrent glioblastoma multiforme. J Clin Neurosci. 2012;19:1636–40. https://doi.org/10.1016/j.jocn.2011.12.028.
van Linde ME, Brahm CG, de Witt Hamer PC, Wagemakers M, Reijneveld JC, Vandertop WP, et al. Survival prediction model of new or progressive intracranial hemorrhage in glioblastoma patients receiving antiangiogenic therapy: a prospective single-center study. J Neurooncol. 2022;159:257–66. https://doi.org/10.1007/s11060-022-04033-0.
Piao Y, Liu N, Xu Y, Li Y, Jiang T, Zhang W, et al. Incidence and risk factors of bleeding in patients with glioblastoma receiving bevacizumab: a retrospective cohort study. J Neurooncol. 2021;155:145–53. https://doi.org/10.1007/s11060-021-03876-z.
Tonder M, Liht M, Stupp R, Taal W. Bevacizumab in glioblastoma: an update on clinical evidence of safety, efficacy and current perspectives. Expert Opin Biol Ther. 2022;22:611–23. https://doi.org/10.1080/14712598.2022.2049182.
Zhou S, Wang L, Li G, Liu R. Factors affecting the incidence of bevacizumab-related thrombocytopenia in Chinese metastatic colorectal cancer patients. Support Care Cancer. 2021;29:4645–53. https://doi.org/10.1007/s00520-021-06007-y.
Plate KH, Scholz A, Dumont DJ. Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited. Acta Neuropathol. 2012;124:763–75. https://doi.org/10.1007/s00401-012-1066-5.
Tsien CI, Pugh SL, Dicker AP, Raizer JJ, Matuszak MM, Lallana EC, et al. NRG oncology/RTOG1205: a randomized phase II trial of concurrent bevacizumab and reirradiation versus bevacizumab alone as treatment for recurrent glioblastoma. J Clin Oncol. 2023;41:1285–95. https://doi.org/10.1200/JCO.22.00164.
Facemire CS, Nixon AB, Griffiths R, Hurwitz H, Coffman TM. Vascular endothelial growth factor receptor 2 controls blood pressure by regulating nitric oxide synthase expression. Hypertension. 2009;54:652–8. https://doi.org/10.1161/HYPERTENSIONAHA.109.133546.
Kappers MH, van Esch JH, Sluiter W, Sleijfer S, Danser AH, van den Meiracker AH. Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 and reduced microvascular density. J Hypertens. 2010;28:2427–37. https://doi.org/10.1097/HJH.0b013e328340d111.
Izzedine H, Ederhy S, Goldwasser F, Soria JC, Milano G, Cohen A, et al. Management of hypertension in angiogenesis inhibitor-treated patients. Ann Oncol. 2009;20:293–300. https://doi.org/10.1093/annonc/mdn573.
van der Veldt AA, Boven E, Helgason HH, van Wouwe M, Berkhof J, de Gast G, et al. Predictive factors for severe toxicity of sunitinib in patients with advanced renal cell cancer. Br J Cancer. 2008;99:259–65. https://doi.org/10.1038/sj.bjc.6604450.
Chen HX, Cleck JN. Adverse effects of anticancer agents that target the VEGF pathway. Nat Rev Clin Oncol. 2009;6:465–77. https://doi.org/10.1038/nrclinonc.2009.94.
Drevs J, Siegert P, Medinger M, Mross K, Strecker R, Zirrgiebel U, et al. Phase I clinical study of AZD2171, an oral vascular endothelial growth factor signaling inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2007;25:3045–54. https://doi.org/10.1200/JCO.2006.07.2066.
Zhu X, Wu S, Dahut WL, Parikh CR. Risks of proteinuria and hypertension with bevacizumab therapy for cancer patients: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2008;3:386–93. https://doi.org/10.2215/CJN.03610807.
Kappers MH, Smedts FM, Horn T, van Esch JH, Sleijfer S, Leijten F, et al. The vascular endothelial growth factor receptor tyrosine kinase inhibitor sunitinib causes a preeclampsia-like syndrome with activation of the endothelin system. Hypertension. 2011;58:295–302. https://doi.org/10.1161/HYPERTENSIONAHA.111.172783.
Arima Y, Oshima S, Noda K, Fukushima H, Taniguchi I, Nakamura S, et al. Sorafenib-induced acute myocardial infarction due to coronary artery spasm. J Cardiol. 2009;54:512–5. https://doi.org/10.1016/j.jjcc.2009.03.009.
Touyz RM, Herrmann J. Cardiotoxicity with vascular endothelial growth factor signaling inhibitors: evidence and mechanisms. Toxicol Pathol. 2018;46:459–71. https://doi.org/10.1177/0192623318781029.
Mancuso MR, Davis R, Norberg SM, O’Brien S, Sennino B, Nakahara T, et al. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J Clin Investig. 2006;116:2610–21. https://doi.org/10.1172/JCI28330.
O’Farrell AM, Abrams TJ, Yuen HA, Ngai TJ, Louie SG, Yee KW, et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood. 2003;101:3597–605. https://doi.org/10.1182/blood-2002-07-2307.
Tsai HT, Marshall JL, Weiss SR, Huang CY, Warren JL, Freedman AN, et al. Bevacizumab use and risk of cardiovascular adverse events among elderly patients with colorectal cancer receiving chemotherapy: a population-based study. Ann Oncol. 2013;24:1574–9. https://doi.org/10.1093/annonc/mdt019.
Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer. 2007;7:475–85. https://doi.org/10.1038/nrc2152.
Iñarrairaegui M, Martinez-Cuesta A, RodrĂguez M, Bilbao JI, Arbizu J, Benito A, et al. Analysis of prognostic factors after yttrium-90 radioembolization of advanced hepatocellular carcinoma. Int J Radiat Oncol Biol Phys. 2010;77:1441–8. https://doi.org/10.1016/j.ijrobp.2009.07.006.
Khorana AA, Connolly GC. Assessing risk of venous thromboembolism in the patient with cancer. J Clin Oncol. 2009;27:4839–47. https://doi.org/10.1200/JCO.2009.22.3271.
Falanga A, Brenner B, Khorana AA, Francis CW. Thrombotic complications in patients with cancer: advances in pathogenesis, prevention, and treatment-A report from ICTHIC 2021. Res Pr Thromb Haemost. 2022;6:e12744. https://doi.org/10.1002/rth2.12744.
Scott BJ, Quant EC, McNamara MB, Ryg PA, Batchelor TT, Wen PY. Bevacizumab salvage therapy following progression in high-grade glioma patients treated with VEGF receptor tyrosine kinase inhibitors. Neuro Oncol. 2010;12:603–7. https://doi.org/10.1093/neuonc/nop073.
Shi Y, Kang X, Ge Y, Cao Y, Li Y, Guo X, et al. The molecular signature and prognosis of glioma with preoperative intratumoral hemorrhage: a retrospective cohort analysis. BMC Neurol. 2024;24:202. https://doi.org/10.1186/s12883-024-03703-2.
Bianconi A, Prior A, Zona G, Fiaschi P. Anticoagulant therapy in high grade gliomas: a systematic review on state of the art and future perspectives. J Neurosurg Sci. 2023;67:236–40. https://doi.org/10.23736/S0390-5616.21.05536-3.
Beal K, Abrey LE, Gutin PH. Antiangiogenic agents in the treatment of recurrent or newly diagnosed glioblastoma: analysis of single-agent and combined modality approaches. Radiat Oncol. 2011;6:2. https://doi.org/10.1186/1748-717X-6-2.
Field KM, Phal PM, Fitt G, Goh C, Nowak AK, Rosenthal MA, et al. The role of early magnetic resonance imaging in predicting survival on bevacizumab for recurrent glioblastoma: Results from a prospective clinical trial (CABARET). Cancer. 2017;123:3576–82. https://doi.org/10.1002/cncr.30838.
Hayes J, French P, Van Den Bent M, Gregory W, Westhead D, Lawler S, et al. Geno-17: an 8-microRNA signature predicts response to bevacizumab in glioblastoma. Neuro Oncol. 2015;17:v95 https://doi.org/10.1093/neuonc/nov215.17.
Kawai N, Watanabe K, Tani N, Sato M, Yamamoto Y, Nishiyama Y, et al. Surgical management and bevacizumab re-challenge in intracranial hemorrhage during glioblastoma therapy: a case report. World Neurosurg. 2023;174:82–88. https://doi.org/10.1016/j.wneu.2023.04.162.
Daneshimehr F, Barabadi Z, Abdolahi S, Soleimani M, Verdi J, Ebrahimi-Barough S, et al. Angiogenesis and its targeting in glioblastoma with focus on clinical approaches. Cell J. 2022;24:555–68. https://doi.org/10.22074/cellj.2022.8154.
Goldman M, Lucke-Wold B, Martinez-Sosa M, Katz J, Mehkri Y, Valisno J, et al. Steroid utility, immunotherapy, and brain tumor management: an update on conflicting therapies. Explor Target Antitumor Ther. 2022;3:659–75. https://doi.org/10.37349/etat.2022.00106.
Giammalva GR, Iacopino DG, Azzarello G, Gaggiotti C, Graziano F, Gulì C, et al. End-of-life care in high-grade glioma patients. The palliative and supportive perspective. Brain Sci. 2018;8:125 https://doi.org/10.3390/brainsci8070125.
Reardon DA, Turner S, Peters KB, Desjardins A, Gururangan S, Sampson JH, et al. A review of VEGF/VEGFR-targeted therapeutics for recurrent glioblastoma. J Natl Compr Cancer Netw. 2011;9:414–27. https://doi.org/10.6004/jnccn.2011.0038.
Castro BA, Aghi MK. Bevacizumab for glioblastoma: current indications, surgical implications, and future directions. Neurosurg Focus. 2014;37:E9 https://doi.org/10.3171/2014.9.focus14516.
Hummel TR, Salloum R, Drissi R, Kumar S, Sobo M, Goldman S, et al. A pilot study of bevacizumab-based therapy in patients with newly diagnosed high-grade gliomas and diffuse intrinsic pontine gliomas. J Neurooncol. 2016;127:53–61. https://doi.org/10.1007/s11060-015-2008-6.
Ammari S, Sallé de Chou R, Assi T, Touat M, Chouzenoux E, Quillent A, et al. Machine-learning-based radiomics MRI model for survival prediction of recurrent glioblastomas treated with bevacizumab. Diagnostics. 2021;11:1263 https://doi.org/10.3390/diagnostics11071263.
Horowitz JR, Rivard A, van der Zee R, Hariawala M, Sheriff DD, Esakof DD, et al. Vascular endothelial growth factor/vascular permeability factor produces nitric oxide-dependent hypotension. Evidence for a maintenance role in quiescent adult endothelium. Arterioscler Thromb Vasc Biol. 1997;17:2793–800. https://doi.org/10.1161/01.atv.17.11.2793.
Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, et al. Glomerular-specific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Investig. 2003;111:707–16. https://doi.org/10.1172/JCI17423.
DeLeve LD, Wang X, Hu L, McCuskey MK, McCuskey RS. Rat liver endothelial cells are targets of LPS-induced oxidative stress. Am J Physiol Gastrointest Liver Physiol. 2004;287:G676–G684. https://doi.org/10.1152/ajpgi.00096.2004.
Kamba T, Tam BY, Hashizume H, Haskell A, Sennino B, Mancuso MR, et al. VEGF-dependent plasticity of fenestrated capillaries in the normal adult microvasculature. Am J Physiol Heart Circ Physiol. 2006;290:H560–76. https://doi.org/10.1152/ajpheart.00133.2005.
Lambrechts D, Carmeliet P. VEGF at the neurovascular interface: therapeutic implications for motor neuron disease?. Biochim Biophys Acta. 2006;1762:1109–21. https://doi.org/10.1016/j.bbadis.2006.09.003.
Inai T, Mancuso M, Hashizume H, Baffert F, Haskell A, Baluk P, et al. Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am J Pathol. 2004;165:35–52. https://doi.org/10.1016/S0002-9440(10)63273-7.
Baffert F, Le T, Sennino B, Thurston G, Kuo CJ, Hu-Lowe D, et al. Cellular changes in normal blood capillaries undergoing regression after inhibition of VEGF signaling. Am J Physiol Heart Circ Physiol. 2006;290:H547–59. https://doi.org/10.1152/ajpheart.00616.2005.
Funding
This work was supported by the Bashkir State Medical University Strategic Academic Leadership Program (PRIORITY-2030).
Author information
Authors and Affiliations
Contributions
Conceptualization, project administration, and writing–original draft, conceptualization, resources, and writing–review and editing, OB and IG; data curation, formal analysis, investigation, and methodology, AK, AA, and VC; software, validation, and visualization, SY and GY; supervision and funding acquisition, OB and IG. All authors agreed on the journal to which the article would be submitted, gave final approval for the version to be published, and agreed to be accountable for all aspects of the work.
Corresponding authors
Ethics declarations
Conflict of interest
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Beylerli, O., Gareev, I., Kaprin, A. et al. Hemorrhagic and ischemic risks of anti-VEGF therapies in glioblastoma. Cancer Gene Ther 32, 762–777 (2025). https://doi.org/10.1038/s41417-025-00914-8
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41417-025-00914-8