Subscribe to RSS
DOI: 10.1055/a-2740-1655
Does the Combination of Anticoagulants and Angiogenesis Inhibitors Increase the Risk of Bleeding in Cancer Patients?
Authors
Supported by: National Natural Science Foundation of China 81973473
Supported by: Natural Science Foundation of Fujian Province 2023J01577
Supported by: Medical Innovation Project of Fujian Province 2024CXA021
Funding Information This work was financially supported by the National Natural Science Foundation of China (81973473); the Natural Science Foundation of Fujian Province (2023J01577); the Medical Innovation Project of Fujian Province (2024CXA021); and the Joint Funds for the Innovation of Science and Technology, Fujian Province (2021Y9152).

Abstract
Background
The advent of angiogenesis inhibitors has expanded therapeutic options for tumors but poses challenges due to bleeding risks, especially in patients requiring anticoagulation therapy for cancer-associated hypercoagulability.
Objectives
This study aimed to evaluate whether combining anticoagulants with angiogenesis inhibitors increases bleeding risk in cancer patients.
Methods
A network meta-analysis was conducted based on PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials to compare bleeding risks with angiogenesis inhibitors alone versus their combination with anticoagulants. Furthermore, a real-world cohort of 645 patients receiving antiangiogenic therapies between January 2010 and June 2024 was studied. Patients were separated into two groups according to whether they were receiving concomitant anticoagulants. The primary outcome was all-grade bleeding events.
Results
Of 2,644 patients from six studies included in network meta-analysis, all-grade bleeding events were found in 614 (23.2%) patients. The addition of anticoagulation to either high-dose bevacizumab (10 or 15 mg/kg) (OR 4.95, 95% CI: 2.68–9.42) or antiangiogenic tyrosine kinase inhibitors (OR 2.2, 95% CI: 1.08–4.44) significantly increased bleeding risk compared with antiangiogenic monotherapy, except for low-dose bevacizumab (5 or 7.5 mg/kg). In the cohort study, 163 patients matched in each group after propensity score matching weighting. Over a median follow-up duration of 56 days, there were 28 (17.2%) all-grade bleeding events during concurrent treatment and 16 (9.8%) all-grade bleeding events reported during antiangiogenic monotherapy.
Conclusion
Adding anticoagulation to high-dose bevacizumab or antiangiogenic TKIs might increase bleeding risk compared with monotherapy. Conversely, anticoagulants appeared to be safe in patients receiving low-dose bevacizumab.
Keywords
angiogenesis inhibitors - anticoagulants - bevacizumab - hemorrhage - tyrosine kinase inhibitors‡ These authors contributed equally to the work.
Publication History
Received: 15 January 2025
Accepted after revision: 05 November 2025
Accepted Manuscript online:
07 November 2025
Article published online:
28 November 2025
© 2025. Thieme. All rights reserved.
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Ferrara N. VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer 2002; 2 (10) 795-803
- 2 Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov 2004; 3 (05) 391-400
- 3 Finn RS, Qin S, Ikeda M. et al; IMbrave150 Investigators. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N Engl J Med 2020; 382 (20) 1894-1905
- 4 Hurwitz H, Fehrenbacher L, Novotny W. et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350 (23) 2335-2342
- 5 Saltz LB, Clarke S, Díaz-Rubio E. et al. Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study. J Clin Oncol 2023; 41: 3663-3669
- 6 Friedman HS, Prados MD, Wen PY. et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 2009; 27 (28) 4733-4740
- 7 Sandler A, Gray R, Perry MC. et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006; 355 (24) 2542-2550
- 8 Burger RA, Brady MF, Bookman MA. et al; Gynecologic Oncology Group. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 2011; 365 (26) 2473-2483
- 9 Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet 2016; 388 (10043): 518-529
- 10 Hirashima M. Regulation of endothelial cell differentiation and arterial specification by VEGF and Notch signaling. Anat Sci Int 2009; 84 (03) 95-101
- 11 Wang S, Li X, Parra M, Verdin E, Bassel-Duby R, Olson EN. Control of endothelial cell proliferation and migration by VEGF signaling to histone deacetylase 7. Proc Natl Acad Sci U S A 2008; 105 (22) 7738-7743
- 12 Kuenen BC, Levi M, Meijers JC. et al. Analysis of coagulation cascade and endothelial cell activation during inhibition of vascular endothelial growth factor/vascular endothelial growth factor receptor pathway in cancer patients. Arterioscler Thromb Vasc Biol 2002; 22 (09) 1500-1505
- 13 Prandoni P, Falanga A, Piccioli A. Cancer and venous thromboembolism. Lancet Oncol 2005; 6 (06) 401-410
- 14 Noble S, Pasi J. Epidemiology and pathophysiology of cancer-associated thrombosis. Br J Cancer 2010; 102 (Suppl. 01) S2-S9
- 15 Abdol Razak NB, Jones G, Bhandari M, Berndt MC, Metharom P. Cancer-associated thrombosis: an overview of mechanisms, risk factors, and treatment. Cancers (Basel) 2018; 10 (10) 380
- 16 Timp JF, Braekkan SK, Versteeg HH, Cannegieter SC. Epidemiology of cancer-associated venous thrombosis. Blood 2013; 122 (10) 1712-1723
- 17 Cinar D, Tas D. Cancer in the elderly. North Clin Istanb 2015; 2 (01) 73-80
- 18 Dunlay SM, Chamberlain AM. Multimorbidity in older patients with cardiovascular disease. Curr Cardiovasc Risk Rep 2016; 10: 3
- 19 Liberati A, Altman DG, Tetzlaff J. et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ 2009; 339: b2700
- 20 Jadad AR, Moore RA, Carroll D. et al. Assessing the quality of reports of randomized clinical trials: is blinding necessary?. Control Clin Trials 1996; 17 (01) 1-12
- 21 Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol 2010; 25 (09) 603-605
- 22 van Valkenhoef G, Dias S, Ades AE, Welton NJ. Automated generation of node-splitting models for assessment of inconsistency in network meta-analysis. Res Synth Methods 2016; 7 (01) 80-93
- 23 Salanti G, Ades AE, Ioannidis JP. Graphical methods and numerical summaries for presenting results from multiple-treatment meta-analysis: an overview and tutorial. J Clin Epidemiol 2011; 64 (02) 163-171
- 24 Veroniki AA, Straus SE, Fyraridis A, Tricco AC. The rank-heat plot is a novel way to present the results from a network meta-analysis including multiple outcomes. J Clin Epidemiol 2016; 76: 193-199
- 25 Higgins JP, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta-analysis: concepts and models for multi-arm studies. Res Synth Methods 2012; 3 (02) 98-110
- 26 Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003; 327 (7414) 557-560
- 27 Schulman S, Kearon C. Subcommittee on Control of Anticoagulation of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis. Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients. J Thromb Haemost 2005; 3 (04) 692-694
- 28 Lin Z, Zhang Q, Luo W. Angiogenesis inhibitors as therapeutic agents in cancer: challenges and future directions. Eur J Pharmacol 2016; 793: 76-81
- 29 Xiao B, Wang W, Zhang D. Risk of bleeding associated with antiangiogenic monoclonal antibodies bevacizumab and ramucirumab: a meta-analysis of 85 randomized controlled trials. OncoTargets Ther 2018; 11: 5059-5074
- 30 Nghiemphu PL, Green RM, Pope WB, Lai A, Cloughesy TF. Safety of anticoagulation use and bevacizumab in patients with glioma. Neuro-oncol 2008; 10 (03) 355-360
- 31 Dansin E, Cinieri S, Garrido P. et al. MO19390 (SAiL): bleeding events in a phase IV study of first-line bevacizumab with chemotherapy in patients with advanced non-squamous NSCLC. Lung Cancer 2012; 76 (03) 373-379
- 32 Reck M, von Pawel J, Zatloukal P. et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol 2009; 27 (08) 1227-1234
- 33 Norden AD, Bartolomeo J, Tanaka S. et al. Safety of concurrent bevacizumab therapy and anticoagulation in glioma patients. J Neurooncol 2012; 106 (01) 121-125
- 34 Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer 2007; 7 (06) 475-485
- 35 Qi WX, Tang LN, Sun YJ. et al. Incidence and risk of hemorrhagic events with vascular endothelial growth factor receptor tyrosine-kinase inhibitors: an up-to-date meta-analysis of 27 randomized controlled trials. Ann Oncol 2013; 24 (12) 2943-2952
- 36 Crist M, Hansen E, Chablani L, Guancial E. Examining the bleeding incidences associated with targeted therapies used in metastatic renal cell carcinoma. Crit Rev Oncol Hematol 2017; 120: 151-162
- 37 Patel SH, George TL, Wang TF. et al. Increased bleeding risk associated with concurrent vascular endothelial growth factor receptor tyrosine kinase inhibitors and low-molecular-weight heparin. Cancer 2021; 127 (06) 938-945
- 38 Sivakumar A, Caulfield SE, Zhang C, Goyal S, Bilen MA, Beardslee TJ. Retrospective study of bleeding risk with concomitant vascular endothelial growth factor receptor tyrosine kinase inhibitor and anticoagulation. Oncologist 2021; 26 (11) e2061-e2069
- 39 Wang TF, Baumann Kreuziger L, Leader A. et al. Characteristics and outcomes of patients on concurrent direct oral anticoagulants and targeted anticancer therapies-TacDOAC registry: communication from the ISTH SSC Subcommittee on Hemostasis and Malignancy. J Thromb Haemost 2021; 19 (08) 2068-2081
- 40 Flaherty KT, Lathia C, Frye RF. et al. Interaction of sorafenib and cytochrome P450 isoenzymes in patients with advanced melanoma: a phase I/II pharmacokinetic interaction study. Cancer Chemother Pharmacol 2011; 68 (05) 1111-1118
- 41 Hussein Z, Mizuo H, Hayato S, Namiki M, Shumaker R. Clinical pharmacokinetic and pharmacodynamic profile of lenvatinib, an orally active, small-molecule, multitargeted tyrosine kinase inhibitor. Eur J Drug Metab Pharmacokinet 2017; 42 (06) 903-914
- 42 Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol 2013; 76 (03) 455-466
- 43 Van der Linden L, Vanassche T, Van Cutsem E, Van Aelst L, Verhamme P. Pharmacokinetic drug-drug interactions with direct anticoagulants in the management of cancer-associated thrombosis. Br J Clin Pharmacol 2023; 89 (08) 2369-2376
- 44 Stevens SM, Woller SC, Baumann Kreuziger L. et al. Antithrombotic therapy for VTE disease: compendium and review of CHEST Guidelines 2012-2021. Chest 2024; 166 (02) 388-404
- 45 Key NS, Khorana AA, Kuderer NM. et al. Venous thromboembolism prophylaxis and treatment in patients with cancer: ASCO clinical practice guideline update. J Clin Oncol 2020; 38 (05) 496-520
- 46 McBane II RD, Wysokinski WE, Le-Rademacher JG. et al. Apixaban and dalteparin in active malignancy-associated venous thromboembolism: the ADAM VTE trial. J Thromb Haemost 2020; 18 (02) 411-421
- 47 Raskob GE, van Es N, Verhamme P. et al; Hokusai VTE Cancer Investigators. Edoxaban for the treatment of cancer-associated venous thromboembolism. N Engl J Med 2018; 378 (07) 615-624
- 48 Agnelli G, Becattini C, Meyer G. et al; Caravaggio Investigators. Apixaban for the treatment of venous thromboembolism associated with cancer. N Engl J Med 2020; 382 (17) 1599-1607
- 49 Denault MH, Morin-Thibault LV, Kermelly SB, Labbé C. Prevention and treatment of thromboembolic events in patients with cancer: advances and challenges. Am J Respir Crit Care Med 2020; 202 (01) 124-127
- 50 Fuentes HE, McBane II RD, Wysokinski WE. et al. Direct oral factor Xa inhibitors for the treatment of acute cancer-associated venous thromboembolism: a systematic review and network meta-analysis. Mayo Clin Proc 2019; 94 (12) 2444-2454
- 51 Huang DQ, Terrault NA, Tacke F. et al. Global epidemiology of cirrhosis—aetiology, trends and predictions. Nat Rev Gastroenterol Hepatol 2023; 20 (06) 388-398
- 52 Huang DQ, Singal AG, Kono Y, Tan DJH, El-Serag HB, Loomba R. Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer. Cell Metab 2022; 34 (07) 969-977.e2
- 53 Anstee QM, Castera L, Loomba R. Impact of non-invasive biomarkers on hepatology practice: past, present and future. J Hepatol 2022; 76 (06) 1362-1378
- 54 Huang DQ, Noureddin N, Ajmera V. et al. Type 2 diabetes, hepatic decompensation, and hepatocellular carcinoma in patients with non-alcoholic fatty liver disease: an individual participant-level data meta-analysis. Lancet Gastroenterol Hepatol 2023; 8 (09) 829-836
- 55 Ajmera V, Cepin S, Tesfai K. et al. A prospective study on the prevalence of NAFLD, advanced fibrosis, cirrhosis and hepatocellular carcinoma in people with type 2 diabetes. J Hepatol 2023; 78 (03) 471-478
- 56 Young AM, Marshall A, Thirlwall J. et al. Comparison of an oral factor Xa inhibitor with low molecular weight heparin in patients with cancer with venous thromboembolism: results of a randomized trial (SELECT-D). J Clin Oncol 2018; 36 (20) 2017-2023
- 57 Frere C, Font C, Esposito F, Crichi B, Girard P, Janus N. Incidence, risk factors, and management of bleeding in patients receiving anticoagulants for the treatment of cancer-associated thrombosis. Support Care Cancer 2022; 30 (04) 2919-2931
- 58 Klok FA, Huisman MV. How I assess and manage the risk of bleeding in patients treated for venous thromboembolism. Blood 2020; 135 (10) 724-734