Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model

Standard

Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model. / Feinauer, Manuel J; Schneider, Stefan W; Berghoff, Anna Sophie; Robador, Jose Ramon; Tehranian, Cedric; Karreman, Matthia A; Venkataramani, Varun; Solecki, Gergely; Grosch, Julia Katharina; Gunkel, Katharina; Kovalchuk, Bogdana; Mayer, Frank Thomas; Fischer, Manuel; Breckwoldt, Michael O; Brune, Maik; Schwab, Yannik; Wick, Wolfgang; Bauer, Alexander Thomas; Winkler, Frank.

In: BLOOD, Vol. 137, No. 9, 04.03.2021, p. 1219-1232.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Feinauer, MJ, Schneider, SW, Berghoff, AS, Robador, JR, Tehranian, C, Karreman, MA, Venkataramani, V, Solecki, G, Grosch, JK, Gunkel, K, Kovalchuk, B, Mayer, FT, Fischer, M, Breckwoldt, MO, Brune, M, Schwab, Y, Wick, W, Bauer, AT & Winkler, F 2021, 'Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model', BLOOD, vol. 137, no. 9, pp. 1219-1232. https://doi.org/10.1182/blood.2020005710

APA

Feinauer, M. J., Schneider, S. W., Berghoff, A. S., Robador, J. R., Tehranian, C., Karreman, M. A., Venkataramani, V., Solecki, G., Grosch, J. K., Gunkel, K., Kovalchuk, B., Mayer, F. T., Fischer, M., Breckwoldt, M. O., Brune, M., Schwab, Y., Wick, W., Bauer, A. T., & Winkler, F. (2021). Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model. BLOOD, 137(9), 1219-1232. https://doi.org/10.1182/blood.2020005710

Vancouver

Feinauer MJ, Schneider SW, Berghoff AS, Robador JR, Tehranian C, Karreman MA et al. Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model. BLOOD. 2021 Mar 4;137(9):1219-1232. https://doi.org/10.1182/blood.2020005710

Bibtex

@article{c99dac4df6f04e38a9b3cc3b03c1aa71,
title = "Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model",
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.",
author = "Feinauer, {Manuel J} and Schneider, {Stefan W} and Berghoff, {Anna Sophie} and Robador, {Jose Ramon} and Cedric Tehranian and Karreman, {Matthia A} and Varun Venkataramani and Gergely Solecki and Grosch, {Julia Katharina} and Katharina Gunkel and Bogdana Kovalchuk and Mayer, {Frank Thomas} and Manuel Fischer and Breckwoldt, {Michael O} and Maik Brune and Yannik Schwab and Wolfgang Wick and Bauer, {Alexander Thomas} and Frank Winkler",
note = "Copyright {\textcopyright} 2020 American Society of Hematology.",
year = "2021",
month = mar,
day = "4",
doi = "10.1182/blood.2020005710",
language = "English",
volume = "137",
pages = "1219--1232",
journal = "BLOOD",
issn = "0006-4971",
publisher = "American Society of Hematology",
number = "9",

}

RIS

TY - JOUR

T1 - Local blood coagulation drives cancer cell arrest and brain metastasis in a mouse model

AU - Feinauer, Manuel J

AU - Schneider, Stefan W

AU - Berghoff, Anna Sophie

AU - Robador, Jose Ramon

AU - Tehranian, Cedric

AU - Karreman, Matthia A

AU - Venkataramani, Varun

AU - Solecki, Gergely

AU - Grosch, Julia Katharina

AU - Gunkel, Katharina

AU - Kovalchuk, Bogdana

AU - Mayer, Frank Thomas

AU - Fischer, Manuel

AU - Breckwoldt, Michael O

AU - Brune, Maik

AU - Schwab, Yannik

AU - Wick, Wolfgang

AU - Bauer, Alexander Thomas

AU - Winkler, Frank

N1 - Copyright © 2020 American Society of Hematology.

PY - 2021/3/4

Y1 - 2021/3/4

N2 - 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.

AB - 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.

U2 - 10.1182/blood.2020005710

DO - 10.1182/blood.2020005710

M3 - SCORING: Journal article

C2 - 33270819

VL - 137

SP - 1219

EP - 1232

JO - BLOOD

JF - BLOOD

SN - 0006-4971

IS - 9

ER -