Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Blood Adv ; 7(11): 2388-2400, 2023 06 13.
Article in English | MEDLINE | ID: mdl-36920782

ABSTRACT

Cancer enhances the risk of venous thromboembolism, but a hypercoagulant microenvironment also promotes cancer progression. Although anticoagulants have been suggested as a potential anticancer treatment, clinical studies on the effect of such modalities on cancer progression have not yet been successful for unknown reasons. In normal physiology, complex formation between the subendothelial-expressed tissue factor (TF) and the blood-borne liver-derived factor VII (FVII) results in induction of the extrinsic coagulation cascade and intracellular signaling via protease-activated receptors (PARs). In cancer, TF is overexpressed and linked to poor prognosis. Here, we report that increased levels of FVII are also observed in breast cancer specimens and are associated with tumor progression and metastasis to the liver. In breast cancer cell lines, tumor-expressed FVII drives changes reminiscent of epithelial-to-mesenchymal transition (EMT), tumor cell invasion, and expression of the prometastatic genes, SNAI2 and SOX9. In vivo, tumor-expressed FVII enhanced tumor growth and liver metastasis. Surprisingly, liver-derived FVII appeared to inhibit metastasis. Finally, tumor-expressed FVII-induced prometastatic gene expression independent of TF but required a functional endothelial protein C receptor, whereas recombinant activated FVII acting via the canonical TF:PAR2 pathway inhibited prometastatic gene expression. Here, we propose that tumor-expressed FVII and liver-derived FVII have opposing effects on EMT and metastasis.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/genetics , Signal Transduction , Thromboplastin/genetics , Thromboplastin/metabolism , Tumor Microenvironment
2.
J Thromb Haemost ; 17(6): 951-963, 2019 06.
Article in English | MEDLINE | ID: mdl-30929299

ABSTRACT

Essentials Factor Xa (FXa)-targeting direct oral anticoagulants (DOACs) reduce venous thromboembolism (VTE) The effects of FXa-targeting DOACs on cancer progression remain to be studied In xenograft models, a FXa-targeting DOAC did not inhibit breast cancer growth and metastasis A thrombin-targeting DOAC, dabigatran, also did not inhibit breast cancer growth and metastasis ABSTRACT: Background Factor Xa-targeting DOACs were recently found to reduce recurrent VTE efficiently in cancer patients when compared to the standard treatment with low-molecular-weight heparins (LMWHs). While the anticancer effects of LMWHs have been extensively studied in preclinical cancer models, the effects of FXa-targeting DOACs on cancer progression remain to be studied. Objective We investigated whether the FXa-targeting DOAC rivaroxaban and the thrombin-targeting DOAC dabigatran etexilate (DE) affected human breast cancer growth and metastasis in orthotopic xenograft models. Methods/results Mice that were put on a custom-made chow diet supplemented with rivaroxaban (0.4 or 1.0 mg/g diet) or dabigatran etexilate (DE) (10 mg/g diet) showed prolonged ex vivo coagulation times (prothrombin time [PT] and activated partial thromboplastin time [aPTT] assay, respectively). However, rivaroxaban and DE did not inhibit MDA-MB-231 tumor growth and metastasis formation in lungs or livers of 7-week-old fully immunodeficient NOD/SCID/Æ´C-/- (NSG) mice. Comparable data were obtained for rivaroxaban-treated mice when using NOD-SCID mice. Rivaroxaban and DE treatment also did not significantly inhibit tumor growth and metastasis formation when using another human triple negative breast cancer (TNBC) cell line (HCC1806) in NOD-SCID mice. The FXa and thrombin-induced gene expression of the downstream target CXCL8 in both cell lines, but FXa and thrombin, did not significantly stimulate migration, proliferation, or stemness in vitro. Conclusion Although effectively inhibiting coagulation, the DOACs rivaroxaban and DE did not inhibit orthotopic growth and metastasis of human TNBC. It remains to be investigated whether DOACs exert antitumorigenic effects in other types of cancer.


Subject(s)
Anticoagulants/pharmacology , Breast Neoplasms/drug therapy , Dabigatran/pharmacology , Rivaroxaban/pharmacology , Animals , Antithrombins/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Factor Xa Inhibitors/pharmacology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Venous Thromboembolism/prevention & control , Xenograft Model Antitumor Assays
3.
Thromb Res ; 140 Suppl 1: S148-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27067969

ABSTRACT

Venous thromboembolism (VTE) and cancer are strongly associated, and present a major challenge in cancer patient treatment. Cancer patients have a higher risk of developing VTE, although the risk differs widely between tumour types. VTE prophylaxis is routinely given to cancer patients, in the form of vitamin K antagonists (VKA) or low molecular weight heparin (LMWH). Several studies have reported that cancer patients receiving anticoagulants show prolonged survival and this effect was more pronounced in patients with a good prognosis, although the mechanism is poorly understood. Tissue Factor (TF) is the initiator of extrinsic coagulation, but its non-haemostatic signalling via protease-activated receptors (PARs) is a potent driver of tumour angiogenesis. Furthermore, coagulation activation is strongly implicated in tumour cell migration and metastasis. This review discusses the effects of anticoagulants on cancer progression in patients, tumour cell behaviour, angiogenesis, and metastasis in in vitro and in vivo models. Inhibition of TF signalling shows great promise in curbing angiogenesis and in vivo tumour growth, but whether this translates to patients is not yet known. Furthermore, non-haemostatic properties of coagulation factors in cancer progression are discussed, which provide exciting opportunities on limiting oncologic processes without affecting blood coagulation.


Subject(s)
Anticoagulants/therapeutic use , Neoplasm Metastasis/prevention & control , Neoplasms/complications , Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Venous Thromboembolism/prevention & control , Animals , Blood Coagulation/drug effects , Coumarins/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Neoplasm Metastasis/pathology , Neoplasms/blood , Neoplasms/pathology , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Thromboplastin/metabolism , Venous Thromboembolism/blood , Venous Thromboembolism/etiology , Venous Thromboembolism/pathology , Vitamin K/antagonists & inhibitors
4.
Thromb Haemost ; 115(5): 1025-33, 2016 05 02.
Article in English | MEDLINE | ID: mdl-26763081

ABSTRACT

Osteosarcoma is the most common primary malignant bone tumour. Patients often develop lung metastasis and have a poor prognosis despite extensive chemotherapy and surgical resections. Tissue Factor is associated with poor clinical outcome in a wide range of cancer types, and promotes angiogenesis and metastasis. The role of Tissue Factor in OS tumourigenesis is unknown. Fifty-three osteosarcoma pre-treatment biopsies and four osteosarcoma cell lines were evaluated for Tissue Factor expression, and a possible association with clinical parameters was investigated. Tissue Factor function was inhibited in an osteosarcoma cell line (143B) by shRNA knockdown or specific antibodies, and pro-tumourigenic gene expression, proliferation, matrigel invasion and transwell migration was examined. 143B cells were implanted in mice in the presence of Tissue Factor-blocking antibodies, and tumour volume, micro-vessel density and metastases in the lung were evaluated. Tissue Factor was highly expressed in 73.6 % of osteosarcoma biopsies, and expression associated significantly with disease-free survival. Tissue Factor was expressed in all four investigated cell lines. Tissue Factor was knocked down in 143B cells, which led to reduced expression of IL-8, CXCL-1, SNAIL and MMP2, but not MMP9. Tissue Factor knockdown or inhibition with antibodies reduced matrigel invasion. Tissue Factor antibodies limited 143B tumour growth in vivo, and resulted in decreased intra-tumoural micro-vessel density. Furthermore, lung metastasis from the primary tumour was significantly reduced. Thus, Tissue Factor expression in osteosarcoma reduces metastasis-free survival in patients, and increases pro-tumourigenic behaviour both in vitro and in vivo.


Subject(s)
Bone Neoplasms/metabolism , Osteosarcoma/metabolism , Thromboplastin/metabolism , Adolescent , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Child , Disease-Free Survival , Female , Gene Expression , Gene Knockdown Techniques , Humans , Male , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Thromboplastin/antagonists & inhibitors , Thromboplastin/genetics , Young Adult
5.
Proc Natl Acad Sci U S A ; 110(28): 11517-22, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23801760

ABSTRACT

Full-length tissue factor (flTF), the coagulation initiator, is overexpressed in breast cancer (BrCa), but associations between flTF expression and clinical outcome remain controversial. It is currently not known whether the soluble alternatively spliced TF form (asTF) is expressed in BrCa or impacts BrCa progression. We are unique in reporting that asTF, but not flTF, strongly associates with both tumor size and grade, and induces BrCa cell proliferation by binding to ß1 integrins. asTF promotes oncogenic gene expression, anchorage-independent growth, and strongly up-regulates tumor expansion in a luminal BrCa model. In basal BrCa cells that constitutively express both TF isoforms, asTF blockade reduces tumor growth and proliferation in vivo. We propose that asTF plays a major role in BrCa progression acting as an autocrine factor that promotes tumor progression. Targeting asTF may comprise a previously unexplored therapeutic strategy in BrCa that stems tumor growth, yet does not impair normal hemostasis.


Subject(s)
Alternative Splicing , Breast Neoplasms/pathology , Integrin beta1/physiology , Thromboplastin/physiology , Adult , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Middle Aged , Thromboplastin/genetics
6.
Genes Chromosomes Cancer ; 49(12): 1095-103, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20737480

ABSTRACT

Conventional osteosarcoma is characterized by rapid growth, high local aggressiveness, and metastasizing potential. Patients developing lung metastases experience poor prognosis despite extensive chemotherapy regimens and surgical interventions. Previously we identified a subgroup of osteosarcoma patients with loss of CDKN2A/p16 protein expression in the primary tumor biopsies which was significantly predictive of a very poor prognosis. Here we aimed to identify the underlying mechanism(s) of this protein loss in relation to osteosarcoma behavior. The CDKN2A locus was analyzed in osteosarcoma cases with total loss of CDKN2A/p16 expression and in cases with high protein expression using melting curve analysis-methylation assay (MCA-Meth), fluorescent in situ hybridization (FISH), multiplex ligation-dependent probe amplification (MLPA), and mutation analysis. All cases with complete CDKN2A/p16 protein loss showed homozygous deletions at the CDKN2A locus. In none of the cases hyper methylation of the promoter region was seen which was confirmed by sequencing this region. Taken together we show that large or smaller deletions of the CDKN2A locus are evident in patient samples and underlie the CDKN2A/p16 protein expression loss while promoter methylation does not appear to be a mechanism of this expression loss. Genomic loss of CDKN2A instead of promoter methylation might be a plausible explanation for the rapid proliferation and high aggressiveness of osteosarcoma by simultaneous impairment CDKN2A/p14(ARF) function.


Subject(s)
Bone Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression , Genes, p16 , Osteosarcoma/genetics , Sequence Deletion , Base Sequence , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Genes, Tumor Suppressor , Humans , In Situ Hybridization, Fluorescence , Prognosis , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...