Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
Blood ; 137(9): 1219-1232, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33270819

ABSTRACT

Clinically relevant brain metastases (BMs) frequently form in cancer patients, with limited options for effective treatment. Circulating cancer cells must first permanently arrest in brain microvessels to colonize the brain, but the critical factors in this process are not well understood. Here, in vivo multiphoton laser-scanning microscopy of the entire brain metastatic cascade allowed unprecedented insights into how blood clot formation and von Willebrand factor (VWF) deposition determine the arrest of circulating cancer cells and subsequent brain colonization in mice. Clot formation in brain microvessels occurred frequently (>95%) and specifically at intravascularly arrested cancer cells, allowing their long-term arrest. An extensive clot embedded ∼20% of brain-arrested cancer cells, and those were more likely to successfully extravasate and form a macrometastasis. Mechanistically, the generation of tissue factor-mediated thrombin by cancer cells accounted for local activation of plasmatic coagulation in the brain. Thrombin inhibition by treatment with low molecular weight heparin or dabigatran and an anti-VWF antibody prevented clot formation, cancer cell arrest, extravasation, and the formation of brain macrometastases. In contrast, tumor cells were not able to directly activate platelets, and antiplatelet treatments did reduce platelet dispositions at intravascular cancer cells but did not reduce overall formation of BMs. In conclusion, our data show that plasmatic coagulation is activated early by intravascular tumor cells in the brain with subsequent clot formation, which led us to discover a novel and specific mechanism that is crucial for brain colonization. Direct or indirect thrombin and VWF inhibitors emerge as promising drug candidates for trials on prevention of BMs.


Subject(s)
Blood Coagulation , Brain Neoplasms/blood , Breast Neoplasms/pathology , Melanoma/pathology , Neoplastic Cells, Circulating/pathology , Thrombosis/blood , Animals , Brain Neoplasms/etiology , Brain Neoplasms/pathology , Breast Neoplasms/blood , Breast Neoplasms/complications , Cell Cycle Checkpoints , Disease Models, Animal , Female , Humans , Melanoma/blood , Melanoma/complications , Mice , Thrombosis/etiology , Thrombosis/pathology , von Willebrand Factor/analysis
3.
Oncotarget ; 7(42): 68527-68545, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27602496

ABSTRACT

Von Willebrand factor (VWF) serves as a nidus for platelet aggregation and thrombosis. We hypothesize that VWF fibers contribute to the development of venous thromboembolism (VTE) and to metastasis formation. Here, we show that vascular and lymphatic endothelial cells (ECs) express VWF in vitro and release VWF fibers after activation by tumor cell supernatants. In contrast, an ex vivo analysis of primary mouse tumors revealed the presence of VWF fibers in the blood microvasculature but not in lymphatic vessels. Unlike the anticoagulant Fondaparinux, an inhibitor of thrombin generation, the low-molecular-weight heparin (LMWH) Tinzaparin inhibited VWF fiber formation and vessel occlusion in tumor vessels by blocking thrombin-induced EC activation and vascular endothelial growth factor-A (VEGF-A)-mediated VWF release. Intradermal tumor cell inoculation in VWF- and ADAMTS13-deficient mice did not alter lymph node metastases compared with wild type animals. Interestingly, multiple tumor-free distal organs exhibited hallmarks of malignancy-related VTE, including luminal VWF fibers, platelet-rich thrombi and vessel occlusions. Furthermore, ADAMTS13 deficiency, characterized by prolonged intraluminal VWF network lifetimes, resulted in a severely increased number of metastatic foci in an experimental model of hematogenous lung seeding. Treatment with Tinzaparin inhibited tumor-induced release of VWF multimers, impeded platelet aggregation and decreased lung metastasis. Thus, our data strongly suggest a critical role of luminal VWF fibers in determining the occurrence of thrombosis and cancer metastasis. Moreover, the findings highlight LMWHs as therapeutic strategy to treat thrombotic complications while executing anti-metastatic activities.


Subject(s)
Heparin, Low-Molecular-Weight/pharmacology , Polysaccharides/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Venous Thromboembolism/prevention & control , von Willebrand Factor/metabolism , Animals , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Factor Xa Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Fondaparinux , Humans , Lymphatic Metastasis , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Tinzaparin , Venous Thromboembolism/genetics , Venous Thromboembolism/metabolism , von Willebrand Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...