组织因子在肿瘤治疗中的应用

王东雁, 李晶, 吴妙芳, 林仲秋

中国实用妇科与产科杂志 ›› 2025, Vol. 41 ›› Issue (9) : 952-956.

PDF(963 KB)
PDF(963 KB)
中国实用妇科与产科杂志 ›› 2025, Vol. 41 ›› Issue (9) : 952-956. DOI: 10.19538/j.fk2025090118
综述与讲座

组织因子在肿瘤治疗中的应用

作者信息 +
文章历史 +

引用本文

导出引用
王东雁, 李晶, 吴妙芳, . 组织因子在肿瘤治疗中的应用[J]. 中国实用妇科与产科杂志. 2025, 41(9): 952-956 https://doi.org/10.19538/j.fk2025090118
中图分类号: R737.3   

参考文献

[1]
Ahmadi SE, Shabannezhad A, Kahrizi A, et al. Tissue factor (coagulation factor Ⅲ):A potential double-edge molecule to be targeted and re-targeted toward cancer[J]. Biomark Res, 2023, 11(1):60. DOI:10.1186/s40364-023-00504-6.
Tissue factor (TF) is a protein that plays a critical role in blood clotting, but recent research has also shown its involvement in cancer development and progression. Herein, we provide an overview of the structure of TF and its involvement in signaling pathways that promote cancer cell proliferation and survival, such as the PI3K/AKT and MAPK pathways. TF overexpression is associated with increased tumor aggressiveness and poor prognosis in various cancers. The review also explores TF's role in promoting cancer cell metastasis, angiogenesis, and venous thromboembolism (VTE). Of note, various TF-targeted therapies, including monoclonal antibodies, small molecule inhibitors, and immunotherapies have been developed, and preclinical and clinical studies demonstrating the efficacy of these therapies in various cancer types are now being evaluated. The potential for re-targeting TF toward cancer cells using TF-conjugated nanoparticles, which have shown promising results in preclinical studies is another intriguing approach in the path of cancer treatment. Although there are still many challenges, TF could possibly be a potential molecule to be used for further cancer therapy as some TF-targeted therapies like Seagen and Genmab’s tisotumab vedotin have gained FDA approval for treatment of cervical cancer. Overall, based on the overviewed studies, this review article provides an in-depth overview of the crucial role that TF plays in cancer development and progression, and emphasizes the potential of TF-targeted and re-targeted therapies as potential approaches for the treatment of cancer.
[2]
Hisada Y, Mackman N. Extracellular Vesicle Tissue Factor Activity Assay[J]. J Vis Exp, 2023, 29:202. DOI: 10.3791/65840.
[3]
Unruh D, Horbinski C. Beyond thrombosis:The impact of tissue factor signaling in cancer[J]. J Hematol Oncol, 2020, 13(1):93. DOI:10.1186/s13045-020-00932-z.
Tissue factor (TF) is the primary initiator of the coagulation cascade, though its effects extend well beyond hemostasis. When TF binds to Factor VII, the resulting TF:FVIIa complex can proteolytically cleave transmembrane G protein-coupled protease-activated receptors (PARs). In addition to activating PARs, TF:FVIIa complex can also activate receptor tyrosine kinases (RTKs) and integrins. These signaling pathways are utilized by tumors to increase cell proliferation, angiogenesis, metastasis, and cancer stem-like cell maintenance. Herein, we review in detail the regulation of TF expression, mechanisms of TF signaling, their pathological consequences, and how it is being targeted in experimental cancer therapeutics.
[4]
Heidari Z, Naeimzadeh Y, Fallahi J, et al. The Role of Tissue Factor In Signaling Pathways of Pathological Conditions and Angiogenesis[J]. Curr Mol Med, 2024, 24(9):1135-1151. DOI:10.2174/0115665240258746230919165935.
Tissue factor (TF) is an integral transmembrane protein associated with the\nextrinsic coagulation pathway. TF gene expression is regulated in response to\ninflammatory cytokines, bacterial lipopolysaccharides, and mechanical injuries. TF\nactivity may be affected by phosphorylation of its cytoplasmic domain and alternative\nsplicing. TF acts as the primary initiator of physiological hemostasis, which prevents\nlocal bleeding at the injury site. However, aberrant expression of TF, accompanied by\nthe severity of diseases and infections under various pathological conditions, triggers\nmultiple signaling pathways that support thrombosis, angiogenesis, inflammation, and\nmetastasis. Protease-activated receptors (PARs) are central in the downstream\nsignaling pathways of TF. In this study, we have reviewed the TF signaling pathways in\ndifferent pathological conditions, such as wound injury, asthma, cardiovascular\ndiseases (CVDs), viral infections, cancer and pathological angiogenesis. Angiogenic\nactivities of TF are critical in the repair of wound injuries and aggressive behavior of\ntumors, which are mainly performed by the actions of vascular endothelial growth\nfactor (VEGF) and hypoxia-inducible factor-1 (HIF1-α). Pro-inflammatory effects of TF\nhave been reported in asthma, CVDs and viral infections, including COVID-19, which\nresult in tissue hypertrophy, inflammation, and thrombosis. TF-FVII induces\nangiogenesis via clotting-dependent and -independent mechanisms. Clottingdependent angiogenesis is induced via the generation of thrombin and cross-linked\nfibrin network, which facilitate vessel infiltration and also act as a reservoir for\nendothelial cells (ECs) growth factors. Expression of TF in tumor cells and ECs\ntriggers clotting-independent angiogenesis through induction of VEGF, urokinase-type\nplasminogen activator (uPAR), early growth response 1 (EGR1), IL8, and cysteine-rich\nangiogenic inducer 61 (Cyr61).
[5]
Hisada Y, Mackman N. Tissue Factor and Cancer: Regulation,Tumor Growth,and Metastasis[J]. Semin Thromb Hemost, 2019, 45(4):385-395. DOI:10.1055/s-0039-1687894.
There is a strong relationship between tissue factor (TF) and cancer. Many cancer cells express high levels of both full-length TF and alternatively spliced (as) TF. TF expression in cancer is associated with poor prognosis. In this review, the authors summarize the regulation of TF expression in cancer cells and the roles of TF and asTF in tumor growth and metastasis. A variety of different signaling pathways, transcription factors and micro ribonucleic acids regulate TF gene expression in cancer cells. The TF/factor VIIa complex enhances tumor growth by activating protease-activated receptor 2 signaling and by increasing the expression of angiogenic factors, such as vascular endothelial growth factor. AsTF increases tumor growth by enhancing integrin β1 signaling. TF and asTF also contribute to metastasis via multiple thrombin-dependent and independent mechanisms that include protecting tumor cells from natural killer cells. Finally, a novel anticancer therapy is using tumor TF as a target to deliver cytotoxic drugs to the tumor. TF may be useful in diagnosis, prognosis, and treatment of cancer.Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.
[6]
de Bono JS, Harris JR, Burm SM, et al. Systematic study of tissue factor expression in solid tumors[J]. Cancer Rep (Hoboken), 2023, 6(2):e1699. DOI:10.1002/cnr2.1699.
Elevated tissue factor (TF) expression, although restricted in normal tissue, has been reported in multiple solid cancers, and expression has been associated with poor prognosis. This manuscript compares TF expression across various solid tumor types via immunohistochemistry in a single study, which has not been performed previously.To increase insight in the prevalence and cellular localization of TF expression across solid cancer types, we performed a detailed and systematic analysis of TF expression in tumor tissue obtained from patients with ovarian, esophageal, bladder, cervical, endometrial, pancreatic, prostate, colon, breast, non-small cell lung cancer (NSCLC), head and neck squamous cell carcinoma (HNSCC), and glioblastoma. The spatial and temporal variation of TF expression was analyzed over time and upon disease progression in patient-matched biopsies taken at different timepoints. In addition, TF expression in patient-matched primary tumor and metastatic lesions was also analyzed.TF expression was detected via immunohistochemistry (IHC) using a validated TF-specific antibody. TF was expressed in all cancer types tested, with highest prevalence in pancreatic cancer, cervical cancer, colon cancer, glioblastoma, HNSCC, and NSCLC, and lowest in breast cancer. Staining was predominantly membranous in pancreatic, cervical, and HNSCC, and cytoplasmic in glioblastoma and bladder cancer. In general, expression was consistent between biopsies obtained from the same patient over time, although variability was observed for individual patients. NSCLC biopsies of primary tumor and matched lymph node metastases showed no clear difference in TF expression overall, although individual patient changes were observed.This study shows that TF is expressed across a broad range of solid cancer types, and expression is present upon tumor dissemination and over the course of treatment.© 2022 The Authors. Cancer Reports published by Wiley Periodicals LLC.
[7]
Carty N, Taylor I, Roath OS, et al. Urinary tissue factor activity in colorectal disease[J]. Br J Surg, 1990, 77(10):1091-1094. DOI:10.1002/bjs.1800771005.
Procoagulant activity (PCA) in normal urine has been recognized for over 50 years. Although tissue factor (TF) is produced by certain tumours, and is increased in both tumour-associated macrophages and blood monocytes, the possibility that it might also be increased in urine has not been studied in patients with cancer. We have measured urinary PCA in hospital controls without inflammatory or neoplastic disease (n = 79), in patients with rheumatoid arthritis (n = 8), inflammatory bowel disease (n = 79), colorectal cancer (n = 70) and in patients undergoing colonoscopy (n = 50). Urinary PCA was higher (P<0.001) in patients with colorectal cancer and inflammatory bowel disease than controls or patients with rheumatoid arthritis. Fourteen (88 per cent) out of 16 colonoscopy patients subsequently found to have carcinoma or inflammatory bowel disease had levels above the control upper quartile, compared with 8 (24 per cent) out of 34 with normal colonoscopy (P < 0.007). TF inhibitors confirmed the nature of the PCA and Western blotting studies indicated a urinary TF molecular weight of approximately 38000. These studies provide further evidence of abnormal haemostasis in malignancy and suggest that determination of urinary TF may provide a useful screening test in patients undergoing colonoscopy.
[8]
Hassan N, Efing J, Kiesel L, et al. The Tissue Factor Pathway in Cancer: Overview and Role of Heparan Sulfate Proteoglycans[J]. Cancers (Basel), 2023, 15(5):1524. DOI:10.3390/cancers15051524.
Historically, the only focus on tissue factor (TF) in clinical pathophysiology has been on its function as the initiation of the extrinsic coagulation cascade. This obsolete vessel-wall TF dogma is now being challenged by the findings that TF circulates throughout the body as a soluble form, a cell-associated protein, and a binding microparticle. Furthermore, it has been observed that TF is expressed by various cell types, including T-lymphocytes and platelets, and that certain pathological situations, such as chronic and acute inflammatory states, and cancer, may increase its expression and activity. Transmembrane G protein-coupled protease-activated receptors can be proteolytically cleaved by the TF:FVIIa complex that develops when TF binds to Factor VII (PARs). The TF:FVIIa complex can activate integrins, receptor tyrosine kinases (RTKs), and PARs in addition to PARs. Cancer cells use these signaling pathways to promote cell division, angiogenesis, metastasis, and the maintenance of cancer stem-like cells. Proteoglycans play a crucial role in the biochemical and mechanical properties of the cellular extracellular matrix, where they control cellular behavior via interacting with transmembrane receptors. For TFPI.fXa complexes, heparan sulfate proteoglycans (HSPGs) may serve as the primary receptor for uptake and degradation. The regulation of TF expression, TF signaling mechanisms, their pathogenic effects, and their therapeutic targeting in cancer are all covered in detail here.
[9]
Wang JG, Geddings JE, Aleman MM, et al. Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer[J]. Blood, 2012, 119(23):5543-5552. DOI:10.1182/blood-2012-01-402156.
Cancer patients often have an activated clotting system and are at increased risk for venous thrombosis. In the present study, we analyzed tissue factor (TF) expression in 4 different human pancreatic tumor cell lines for the purpose of producing derivative tumors in vivo. We found that 2 of the lines expressed TF and released TF-positive microparticles (MPs) into the culture medium. The majority of TF protein in the culture medium was associated with MPs. Only TF-positive cell lines activated coagulation in nude mice, and this activation was abolished by an anti–human TF Ab. Of the 2 TF-positive lines, only one produced detectable levels of human MP TF activity in the plasma when grown orthotopically in nude mice. Surprisingly, < 5% of human TF protein in plasma from tumor-bearing mice was associated with MPs. Mice with TF-positive tumors and elevated levels of circulating TF-positive MPs had increased thrombosis in a saphenous vein model. In contrast, we observed no difference in thrombus weight between tumor-bearing and control mice in an inferior vena cava stenosis model. The results of the present study using a xenograft mouse model suggest that tumor TF activates coagulation, whereas TF on circulating MPs may trigger venous thrombosis.
[10]
Rees PA, Castle J, Clouston HW, et al. The effects of coagulation factors and their inhibitors on proliferation and migration in colorectal cancer[J]. Cancer Med, 2023, 12(16):17184-17192. DOI:10.1002/cam4.6332.
Clotting factors promote cancer development. We investigated if coagulation proteins promote proliferation and migration in colorectal cancer (CRC) cell lines and whether their direct inhibitors can attenuate these effects.
[11]
Zhang J, Ding J, Zhang X, et al. Regulation of vascular endothelial growth factor (VEGF) production and angiogenesis by tissue Factor (TF) in SGC-7901 gastric cancer cells[J]. Cancer Biol Ther, 2005, 4(7):769-772. DOI:10.4161/cbt.4.7.1871.
Tissue factor (TF), an initiator of the extrinsic coagulation cascade, is expressed in a wide range of cancer cells and plays important roles in cancer progression and metastasis. We demonstrated between TF and vascular endothelial growth factor (VEGF) production differences in four human gastric cell lines. One of these cell lines, SGC-7901, a high TF and VEGF producer, was grown subcutaneously in severe combined immuno-deficient (SCID) mice. The SCID mice generated solid tumors characterized by intense vascularity. In contrast, SGC-7901 cells transfected with antisense TF cDNA generated relatively avascular tumors in SCID mice, as determined by immunohistochemical staining of tumor vascular endothelial cells with anti-VIII factor antibody. To investigate the structure-function relationship between TF and VEGF, the SGC-7901 cell line was transfected with antisense a full-length TF cDNA, a cytoplasmic deletion mutant lacking the distal three serine residues (potential substrates for protein kinase C), or an extracellular domain mutant, which has markedly diminished function for activation of factor X. Cells transfected with the full-length antisense TF sequence produced decreased levels of both TF and VEGF. Transfectants with the extracellular domain mutant produced high levels of VEGF mRNA. However, cells transfected with the cytoplasmic deletion mutant construct produced increased levels of TF, but little or no VEGF. Thus, the cytoplasmic tail of TF may signal VEGF expression in some tumor cells.
[12]
Matiash K, Lewis CS, Bogdanov VY. Functional Characteristics and Regulated Expression of Alternatively Spliced Tissue Factor: An Update[J]. Cancers (Basel), 2021, 13(18):4652. DOI:10.3390/cancers13184652.
In human and mouse, alternative splicing of tissue factor’s primary transcript yields two mRNA species: one features all six TF exons and encodes full-length tissue factor (flTF), and the other lacks exon 5 and encodes alternatively spliced tissue factor (asTF). flTF, which is oftentimes referred to as “TF”, is an integral membrane glycoprotein due to the presence of an alpha-helical domain in its C-terminus, while asTF is soluble due to the frameshift resulting from the joining of exon 4 directly to exon 6. In this review, we focus on asTF—the more recently discovered isoform of TF that appears to significantly contribute to the pathobiology of several solid malignancies. There is currently a consensus in the field that asTF, while dispensable to normal hemostasis, can activate a subset of integrins on benign and malignant cells and promote outside-in signaling eliciting angiogenesis; cancer cell proliferation, migration, and invasion; and monocyte recruitment. We provide a general overview of the pioneering, as well as more recent, asTF research; discuss the current concepts of how asTF contributes to cancer progression; and open a conversation about the emerging utility of asTF as a biomarker and a therapeutic target.
[13]
Milsom CC, Yu JL, Mackman N, et al. Tissue factor regulation by epidermal growth factor receptor and epithelial-to-mesenchymal transitions:Effect on tumor initiation and angiogenesis[J]. Cancer Res, 2008, 68(24):10068-10076. DOI:10.1158/0008-5472.CAN-08-2067.
ErbB oncogenes drive the progression of several human cancers. Our study shows that in human carcinoma (A431) and glioma (U373) cells, the oncogenic forms of epidermal growth factor receptor (EGFR; including EGFRvIII) trigger the up-regulation of tissue factor (TF), the transmembrane protein responsible for initiating blood coagulation and signaling through interaction with coagulation factor VIIa. We show that A431 cancer cells in culture exhibit a uniform TF expression profile; however, these same cells in vivo exhibit a heterogeneous TF expression and show signs of E-cadherin inactivation, which is coupled with multilineage (epithelial and mesenchymal) differentiation. Blockade of E-cadherin in vitro, leads to the acquisition of spindle morphology and de novo expression of vimentin, features consistent with epithelial-to-mesenchymal transition. These changes were associated with an increase in EGFR-dependent TF expression, and with enhanced stimulation of vascular endothelial growth factor production, particularly following cancer cell treatment with coagulation factor VIIa. In vivo, cells undergoing epithelial-to-mesenchymal transition exhibited an increased metastatic potential. Furthermore, injections of the TF-blocking antibody (CNTO 859) delayed the initiation of A431 tumors in immunodeficient mice, and reduced tumor growth, vascularization, and vascular endothelial growth factor expression. Collectively, our data suggest that TF is regulated by both oncogenic and differentiation pathways, and that it functions in tumor initiation, tumor growth, angiogenesis, and metastasis. Thus, TF could serve as a therapeutic target in EGFR-dependent malignancies.
[14]
Li H, Yu Y, Gao L, et al. Tissue factor:A neglected role in cancer biology[J]. J Thromb Thrombolysis, 2022, 54(1):97-108. DOI:10.1007/s11239-022-02662-0.
[15]
Shaker H, Harrison H, Clarke R, et al. Tissue Factor promotes breast cancer stem cell activity in vitro[J]. Oncotarget, 2017, 8(16):25915-25927. DOI:10.18632/oncotarget.13928.
Cancer stem cells (CSCs) are a subpopulation of cells that can self-renew and initiate tumours. The clotting-initiating protein Tissue Factor (TF) promotes metastasis and may be overexpressed in cancer cells with increased CSC activity. We sought to determine whether TF promotes breast CSC activity in vitro using human breast cancer cell lines. TF expression was compared in anoikis-resistant (CSC-enriched) and unselected cells. In cells sorted into of TF-expressing and TF-negative (FACS), and in cells transfected to knockdown TF (siRNA) and overexpress TF (cDNA), CSC activity was compared by (i) mammosphere forming efficiency (MFE) (ii) holoclone colony formation (Hc) and (iii) ALDH1 activity. TF expression was increased in anoikis-resistant and high ALDH1-activity T47D cells compared to unselected cells. FACS sorted TF-expressing T47Ds and TF-overexpressing MCF7s had increased CSC activity compared to TF-low cells. TF siRNA cells (MDAMB231,T47D) had reduced CSC activity compared to control cells. FVIIa increased MFE and ALDH1 in a dose-dependent manner (MDAMB231, T47D). The effects of FVIIa on MFE were abrogated by TF siRNA (T47D). Breast CSCs (in vitro) demonstrate increased activity when selected for high TF expression, when induced to overexpress TF, and when stimulated (with FVIIa). Targeting the TF pathway in vivo may abrogate CSC activity.
[16]
Clouston HW, Rees PA, Lamb R, et al. Effect of Tissue Factor on Colorectal Cancer Stem Cells[J]. Anticancer Res, 2018, 38(5):2635-2642. DOI: 10.21873/anticanres.
Tissue factor (TF) expression increases cancer stem cell (CSC) activity in breast and lung cancer. There are ongoing studies focused on targeting CSCs via anti-TF treatment, for breast and lung cancer therapy. Herein, the aim was to determine whether targeting TF could have an anti-CSC therapeutic role in colorectal cancer (CRC).Evaluation of colonosphere-forming efficiency (CFE) and aldehyde dehydrogenase (ALDH) expression level was used to quantify CSC activity in two CRC cell lines, after TF knockdown (TFKD) or TF over-expression (TFOE).TFKD resulted in increased levels of ALDH in SW620 (1.31±0.04-fold, p<0.001) and DLD-1 (1.63±0.14-fold, p=0.04) cells. CFE was increased in SW620 (1.21±0.23% vs. 2.03±0.29%, p=0.01) and DLD-1 (0.41±0.12% vs. 0.68±0.9%, p=0.01) cells. Conversely, TFOE decreased ALDH expression (0.72±0.04-fold, p=0.001) and CFE (0.33±0.05% vs. 0.66±0.14%, p=0.006) in DLD-1, but had no impact on SW620 cells.In the examined CRC cell lines, TF expression was inversely related to CSC activity suggesting that anti-TF therapies may not have a role in CRC treatment.Copyright© 2018, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.
[17]
Bourcy M, Suarez-Carmona M, Lambert J, et al. Tissue Factor Induced by Epithelial-Mesenchymal Transition Triggers a Procoagulant State That Drives Metastasis of Circulating Tumor Cells[J]. Cancer Res, 2016, 76(14):4270-4282. DOI:10.1158/0008-5472.CAN-15-2263.
Epithelial-mesenchymal transition (EMT) is prominent in circulating tumor cells (CTC), but how it influences metastatic spread in this setting is obscure. Insofar as blood provides a specific microenvironment for tumor cells, we explored a potential link between EMT and coagulation that may provide EMT-positive CTCs with enhanced colonizing properties. Here we report that EMT induces tissue factor (TF), a major cell-associated initiator of coagulation and related procoagulant properties in the blood. TF blockade by antibody or shRNA diminished the procoagulant activity of EMT-positive cells, confirming a functional role for TF in these processes. Silencing the EMT transcription factor ZEB1 inhibited both EMT-associated TF expression and coagulant activity, further strengthening the link between EMT and coagulation. Accordingly, EMT-positive cells exhibited a higher persistance/survival in the lungs of mice colonized after intravenous injection, a feature diminished by TF or ZEB1 silencing. In tumor cells with limited metastatic capability, enforcing expression of the EMT transcription factor Snail increased TF, coagulant properties, and early metastasis. Clinically, we identified a subpopulation of CTC expressing vimentin and TF in the blood of metastatic breast cancer patients consistent with our observations. Overall, our findings define a novel EMT-TF regulatory axis that triggers local activation of coagulation pathways to support metastatic colonization of EMT-positive CTCs. Cancer Res; 76(14); 4270-82. ©2016 AACR.©2016 American Association for Cancer Research.
[18]
Li X, Cao D, Zheng X, et al. Tissue factor as a new target for tumor therapy-killing two birds with one stone:A narrative review[J]. Ann Transl Med, 2022, 10(22):1250. DOI:10.21037/atm-22-5067.
[19]
Arnault JP, Chemmama K, Ferroudj K, et al. The Dynamic Landscape of the Coagulome of Metastatic Malignant Melanoma[J]. Int J Mol Sci, 2025, 26(4):1435. DOI:10.3390/ijms26041435.
The local expression of coagulation-related genes defines the tumor coagulome. The tumor coagulome plays a pivotal role in cancer-associated thrombosis (CAT) and hemostatic complications, such as venous thromboembolism (VTE), which are frequent in patients with advanced/metastatic cancer. Genomic analyses of human tumors, such as skin cutaneous melanoma (SKCM), have unveiled the complexity of the metastatic trajectories. However, no study to date has focused on the metastatic coagulome along these trajectories. Using bulk-tumor and single-cell analyses of primary SKCM, metastastic samples and circulating tumor cells (CTCs), we explored the coagulome of SKCM along metastatic progression. We identified consistent changes in the coagulome of SKCM metastases compared to primary tumors and observed metastatic site specificity. Compared to other metastatic sites, lung metastases of SKCM had a specific coagulome with a higher expression of F3, encoding Tissue Factor. Single-cell analyses were used to chart the inter- and intra-tumor heterogeneity and characterize the metastatic coagulome of SKCM. We found that a subpopulation of CTCs from SKCM expressed high levels of platelet genes, suggesting the contribution of CTC–platelet interactions to the CTC coagulome. These findings highlight the dynamic properties of the metastatic coagulome and its link to cancer progression.
[20]
Wang X, Wang M, Amarzguioui M, et al. Downregulation of tissue factor by RNA interference in human melanoma LOX-L cells reduces pulmonary metastasis in nude mice[J]. Int J Cancer, 2004, 112(6):994-1002. DOI: 10.1002/ijc.20527.
Tissue factor (TF) is the membrane receptor of the serine protease coagulation factor VIIa (FVIIa). Formation of the TF/FVIIa complex initiates the coagulation cascade. We used short hairpin RNA (shRNA)-mediated RNA interference to knock down TF expression in the human metastatic melanoma cell line LOX-L. After transfection with the shRNA construct, 3 stable clones with significantly downregulated TF expression were established. They exhibited decreased proliferation in vitro as determined by (14)C thymidine incorporation and soft agar assay. The in vivo metastatic potential was assessed in an experimental pulmonary metastasis model in which cells from different clones were injected into the tail vein of nude mice. The incidence of pulmonary tumors was significantly lower in mice receiving shRNA-expressing cells (33% +/- 15%) than in control mice injected with wild-type cells or cells stably transfected with empty expression vector (90% +/- 10%). The mice injected with TF-downregulated cells had markedly longer survival time (69 +/- 17 days) compared to the control mice (35.6 +/- 5 days; p = 0.03). Thus, reduction of TF levels in LOX-L cells significantly delayed and reduced lung tumor formation. As a first step in elucidating the molecular basis for this effect, we compared the global gene expression profile in TF-downregulated cells and control cells by using cDNA microarray analysis. Forty-four known human genes were found to be significantly upregulated (> 2-fold; p < 0.05) and 228 genes significantly downregulated (>or= 3-fold; p < 0.05) in TF-downregulated cells compared to control cells. The differentially expressed genes encode proteins functioning in transcription, translation, cell communication and cell growth/death. The results provide a basis for investigating molecular mechanisms underlying the effects of TF on the metastatic capacity of LOX-L melanoma cells.(c) 2004 Wiley-Liss, Inc.
[21]
Versteeg HH, Schaffner F, Kerver M, et al. Inhibition of tissue factor signaling suppresses tumor growth[J]. Blood, 2008, 111(1):190-199. DOI: 10.1182/blood-2007-07-101048.
Coagulation activation by tissue factor (TF) is implicated in cancer progression, cancer-associated thrombosis and metastasis. The role of direct TF signaling pathways in cancer, however, remains incompletely understood. Here we address how TF contributes to primary tumor growth by using a unique pair of isotype-matched antibodies that inhibit either coagulation (monoclonal antibody [Mab]-5G9) or direct signaling (Mab-10H10). We demonstrate that the inhibitory antibody of direct TF-VIIa signaling not only blocks TF-VIIa mediated activation of PAR2, but also disrupts the interaction of TF with integrins. In epithelial and TF-expressing endothelial cells, association of TF with beta1 integrins is regulated by TF extracellular ligand binding and independent of PAR2 signaling or proteolytic activity of VIIa. In contrast, alpha3beta1 integrin association of TF is constitutive in breast cancer cells and blocked by Mab-10H10 but not by Mab-5G9. Mab-5G9 has antitumor activity in vivo, but we show here that Mab-10H10 is at least as effective in suppressing human xenograft tumors in 2 different models. Breast tumor growth was also attenuated by blocking PAR2 signaling. These results show that tumor cell TF-PAR2 signaling is crucial for tumor growth and suggest that anti-TF strategies can be applied in cancer therapy with minor impairment of TF-dependent hemostatic pathways.
[22]
de Goeij BE, Satijn D, Freitag CM, et al. High turnover of tissue factor enables efficient intracellular delivery of antibody-drug conjugates[J]. Mol Cancer Ther, 2015, 14(5):1130-1140. DOI:10.1158/1535-7163.MCT-14-0798.
Antibody-drug conjugates (ADC) are emerging as powerful cancer treatments that combine antibody-mediated tumor targeting with the potent cytotoxic activity of toxins. We recently reported the development of a novel ADC that delivers the cytotoxic payload monomethyl auristatin E (MMAE) to tumor cells expressing tissue factor (TF). By carefully selecting a TF-specific antibody that interferes with TF:FVIIa-dependent intracellular signaling, but not with the procoagulant activity of TF, an ADC was developed (TF-011-MMAE/HuMax-TF-ADC) that efficiently kills tumor cells, with an acceptable toxicology profile. To gain more insight in the efficacy of TF-directed ADC treatment, we compared the internalization characteristics and intracellular routing of TF with the EGFR and HER2. Both in absence and presence of antibody, TF demonstrated more efficient internalization, lysosomal targeting, and degradation than EGFR and HER2. By conjugating TF, EGFR, and HER2-specific antibodies with duostatin-3, a toxin that induces potent cytotoxicity upon antibody-mediated internalization but lacks the ability to induce bystander killing, we were able to compare cytotoxicity of ADCs with different tumor specificities. TF-ADC demonstrated effective killing against tumor cell lines with variable levels of target expression. In xenograft models, TF-ADC was relatively potent in reducing tumor growth compared with EGFR- and HER2-ADCs. We hypothesize that the constant turnover of TF on tumor cells makes this protein specifically suitable for an ADC approach.©2015 American Association for Cancer Research.
[23]
Hong DS, Concin N, Vergote I, et al. Tisotumab Vedotin in Previously Treated Recurrent or Metastatic Cervical Cancer[J]. Clin Cancer Res, 2020, 26(6):1220-1228. DOI: 10.1158/1078-0432.CCR-19-2962.
Tissue factor (TF) is a potential target in cervical cancer, as it is frequently highly expressed and associated with poor prognosis. Tisotumab vedotin, a first-in-class investigational antibody-drug conjugate targeting TF, has demonstrated encouraging activity in solid tumors. Here we report data from the cervical cancer cohort of innovaTV 201 phase I/II study (NCT02001623).Patients with recurrent or metastatic cervical cancer received tisotumab vedotin 2.0 mg/kg every 3 weeks until progressive disease, unacceptable toxicity, or consent withdrawal. The primary objective was safety and tolerability. Secondary objectives included antitumor activity.Of the 55 patients, 51% had received ≥2 prior lines of treatment in the recurrent or metastatic setting; 67% had prior bevacizumab + doublet chemotherapy. Fifty-one percent of patients had squamous cell carcinoma. The most common grade 3/4 treatment-emergent adverse events (AEs) were anemia (11%), fatigue (9%), and vomiting (7%). No grade 5 treatment-related AEs occurred. Investigator-assessed confirmed objective response rate (ORR) was 24% [95% confidence interval (CI): 13%-37%]. Median duration of response (DOR) was 4.2 months (range: 1.0-9.7); four patients responded for >8 months. The 6-month progression-free survival (PFS) rate was 29% (95% CI: 17%-43%). Independent review outcomes were comparable, with confirmed ORR of 22% (95% CI: 12%-35%), median DOR of 6.0 months (range: 1.0-9.7), and 6-month PFS rate of 40% (95% CI: 24%-55%). Tissue factor expression was confirmed in most patients; no significant association with response was observed.Tisotumab vedotin demonstrated a manageable safety profile and encouraging antitumor activity in patients with previously treated recurrent or metastatic cervical cancer.©2019 American Association for Cancer Research.
[24]
Coleman RL, Lorusso D, Gennigens C, et al. Efficacy and safety of tisotumab vedotin in previously treated recurrent or metastatic cervical cancer (innovaTV 204/GOG-3023/ENGOT-cx6):A multicentre,open-label,single-arm,phase 2 study[J]. Lancet Oncol, 2021, 22(5):609-619. DOI:10.1016/S1470-2045(21)00056-5.
Few effective second-line treatments exist for women with recurrent or metastatic cervical cancer. Accordingly, we aimed to evaluate the efficacy and safety of tisotumab vedotin, a tissue factor-directed antibody-drug conjugate, in this patient population.This multicentre, open-label, single-arm, phase 2 study was done across 35 academic centres, hospitals, and community practices in Europe and the USA. The study included patients aged 18 years or older who had recurrent or metastatic squamous cell, adenocarcinoma, or adenosquamous cervical cancer; disease progression on or after doublet chemotherapy with bevacizumab (if eligible by local standards); who had received two or fewer previous systemic regimens for recurrent or metastatic disease; had measurable disease based on Response Evaluation Criteria in Solid Tumors (RECIST; version 1.1); and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients received 2·0 mg/kg (up to a maximum of 200 mg) tisotumab vedotin intravenously once every 3 weeks until disease progression (determined by the independent review committee) or unacceptable toxicity. The primary endpoint was confirmed objective response rate based on RECIST (version 1.1), as assessed by the independent review committee. Activity and safety analyses were done in patients who received at least one dose of the drug. This study is ongoing with recruitment completed and is registered with ClinicalTrials.gov, NCT03438396.102 patients were enrolled between June 12, 2018, and April 11, 2019; 101 patients received at least one dose of tisotumab vedotin. Median follow-up at the time of analysis was 10·0 months (IQR 6·1-13·0). The confirmed objective response rate was 24% (95% CI 16-33), with seven (7%) complete responses and 17 (17%) partial responses. The most common treatment-related adverse events included alopecia (38 [38%] of 101 patients), epistaxis (30 [30%]), nausea (27 [27%]), conjunctivitis (26 [26%]), fatigue (26 [26%]), and dry eye (23 [23%]). Grade 3 or worse treatment-related adverse events were reported in 28 (28%) patients and included neutropenia (three [3%] patients), fatigue (two [2%]), ulcerative keratitis (two [2%]), and peripheral neuropathies (two [2%] each with sensory, motor, sensorimotor, and neuropathy peripheral). Serious treatment-related adverse events occurred in 13 (13%) patients, the most common of which included peripheral sensorimotor neuropathy (two [2%] patients) and pyrexia (two [2%]). One death due to septic shock was considered by the investigator to be related to therapy. Three deaths unrelated to treatment were reported, including one case of ileus and two unknown causes.Tisotumab vedotin showed clinically meaningful and durable antitumour activity with a manageable and tolerable safety profile in women with previously treated recurrent or metastatic cervical cancer. Given the poor prognosis for this patient population and the low activity of current therapies in this setting, tisotumab vedotin, if approved, would represent a new treatment for women with recurrent or metastatic cervical cancer.Genmab, Seagen, Gynaecologic Oncology Group, and European Network of Gynaecological Oncological Trial Groups.Copyright © 2021 Elsevier Ltd. All rights reserved.
[25]
周晖, 刘昀昀, 余孝丽, 等. 《2025 NCCN子宫颈癌临床实践指南(第1版)》解读[J]. 中国实用妇科与产科杂志, 2025, 41(1):96-102. DOI:10.19538/j.fk2025010122.
[26]
Vergote I, González-Martín A, Fujiwara K, et al. Tisotumab Vedotin as Second- or Third-Line Therapy for Recurrent Cervical Cancer[J]. N Engl J Med, 2024, 391(1):44-55. DOI:10.1056/NEJMoa2313811.
[27]
Vergote I, Van Nieuwenhuysen E, O'Cearbhaill RE, et al. Tisotumab Vedotin in Combination With Carboplatin,Pembrolizumab,or Bevacizumab in Recurrent or Metastatic Cervical Cancer: Results From the innovaTV 205/GOG-3024/ENGOT-cx8 Study[J]. J Clin Oncol, 2023, 41(36):5536-5549. DOI:10.1200/JCO.23.00720.
[28]
Sun L, Fayette L, Salas S, et al. Tisotumab vedotin in head and neck squamous cell carcinoma:Updated analysis from innovaTV 207 Part C[J]. J Clin Oncol, 2024, 42(suppl 16): 6012.
6012
[29]
中国抗癌协会宫颈癌专业委员会, 周晖, 张丙忠, 等. 妇科恶性肿瘤抗体偶联药物临床应用指南(2024年版)[J]. 中国实用妇科与产科杂志, 2024, 40(5):516-525. DOI:10.19538/j.fk2024050111.
[30]
Full prescribing information for TIVDAK[EB/OL]. (2025-02-05)[2025-06-01]. https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/761208s007lbl.pdf.
[31]
崔广霞, 陈麟凤, 白文佩, 等. 妇科恶性肿瘤围术期患者血液管理专家共识(2025年版)[J]. 中国实用妇科与产科杂志, 2025, 41(3):316-323. DOI:10.19538/j.fk2025030113.
[32]
于仲勋, 高思豪, 马明圣. Ⅰ型干扰素病研究进展[J]. 中国实用儿科杂志, 2024, 39(6):444-448. DOI:10.19538/j.ek2024060608.
[33]
卢淮武, 叶栋栋, 吴斌, 等. 《2023 NCCN卵巢癌包括输卵管癌及原发性腹膜癌临床实践指南(第1版)》解读[J]. 中国实用妇科与产科杂志, 2023, 39(1):58-67. DOI:10.19538/j.fk2023010116.

基金

国家自然科学基金(82403264)
广东省基础与应用基础研究基金(2023A1515012647)
广州市临床重大技术项目(2023P-ZD12)

PDF(963 KB)

Accesses

Citation

Detail

段落导航
相关文章

/