1
|
Kayser A, Wolff A, Berlin P, Duehring L, Henze L, Mundkowski R, Bergmann W, Müller-Hilke B, Wagner C, Huehns M, Oehmcke-Hecht S, Maletzki C. Selective but not pan-CDK inhibition abrogates 5-FU-driven tissue factor upregulation in colon cancer. Sci Rep 2024; 14:10582. [PMID: 38719932 PMCID: PMC11078971 DOI: 10.1038/s41598-024-61076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Thromboembolic events are complications in cancer patients and hypercoagulability has been linked to the tissue factor (TF) pathway, making this an attractive target. Here, we investigated the effects of chemotherapeutics and CDK inhibitors (CDKI) abemaciclib/palbociclib (CDK4/6), THZ-1 (CDK7/12/13), and dinaciclib (CDK1/2/5/9) alone and in combination regimens on TF abundance and coagulation. The human colorectal cancer (CRC) cell line HROC173 was treated with 5-FU or gemcitabine to stimulate TF expression. TF+ cells were sorted, recultured, and re-analyzed. The effect of treatment alone or in combination was assessed by functional assays. Low-dose chemotherapy induced a hypercoagulable state and significantly upregulated TF, even after reculture without treatment. Cells exhibited characteristics of epithelial-mesenchymal transition, including high expression of vimentin and mucin. Dinaciclib and THZ-1 also upregulated TF, while abemaciclib and palbociclib downregulated it. Similar results were observed in coagulation assays. The same anticoagulant activity of abemaciclib was seen after incubation with peripheral immune cells from healthy donors and CRC patients. Abemaciclib reversed 5-FU-induced TF upregulation and prolonged clotting times in second-line treatment. Effects were independent of cytotoxicity, senescence, and p27kip1 induction. TF-antibody blocking experiments confirmed the importance of TF in plasma coagulation, with Factor XII playing a minor role. Short-term abemaciclib counteracts 5-FU-induced hypercoagulation and eventually even prevents thromboembolic events.
Collapse
Affiliation(s)
- Annika Kayser
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Annabell Wolff
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany
| | - Peggy Berlin
- Department of Medicine II, Division of Gastroenterology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Lara Duehring
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany
| | - Larissa Henze
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
- Department of Internal Medicine II, Asklepios Hospital Harz, Goslar, Germany
| | - Ralf Mundkowski
- Center of Pharmacology and Toxicology, Institute of Clinical Pharmacology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Wendy Bergmann
- Laboratory for Clinical Immunology, Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057, Rostock, Germany
| | - Brigitte Müller-Hilke
- Laboratory for Clinical Immunology, Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057, Rostock, Germany
| | - Charlotte Wagner
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Maja Huehns
- Institute of Pathology, Rostock University Medical Center, 18057, Rostock, Germany
| | - Sonja Oehmcke-Hecht
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057, Rostock, Germany.
| | - Claudia Maletzki
- Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
2
|
Majood M, Rawat S, Mohanty S. Delineating the role of extracellular vesicles in cancer metastasis: A comprehensive review. Front Immunol 2022; 13:966661. [PMID: 36059497 PMCID: PMC9439583 DOI: 10.3389/fimmu.2022.966661] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/01/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are subcellular messengers that aid in the formation and spread of cancer by enabling tumor-stroma communication. EVs develop from the very porous structure of late endosomes and hold information on both the intrinsic “status” of the cell and the extracellular signals absorbed by the cells from their surroundings. These EVs contain physiologically useful components, including as nucleic acids, lipids, and proteins, which have been found to activate important signaling pathways in tumor and tumor microenvironment (TME) cells, aggravating tumor growth. We highlight critical cell biology mechanisms that link EVS formation to cargo sorting in cancer cells in this review.Sorting out the signals that control EVs creation, cargo, and delivery will aid our understanding of carcinogenesis. Furthermore, we reviewed how cancer development and spreading behaviors are affected by coordinated communication between malignant and non-malignant cells. Herein, we studied the reciprocal exchanges via EVs in various cancer types. Further research into the pathophysiological functions of various EVs in tumor growth is likely to lead to the discovery of new biomarkers in liquid biopsy and the development of tumor-specific therapies.
Collapse
|
3
|
Fillmann LS, Fillmann LP, Oliveira LZD, Fillmann HS, Carvalhal GF. Tissue Factor Expression in Colorectal Adenocarcinoma: Association with Angiogenesis and Clinical and Pathological Aspects. JOURNAL OF COLOPROCTOLOGY 2022. [DOI: 10.1055/s-0041-1739453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Abstract
Introduction Tissue factor (TF) expression has been described in various neoplasms and was correlated with angiogenesis and metastases.
Objectives To describe TF expression in colorectal cancers, correlating it with microvessel density and clinical and pathological variables.
Methods Immunohistochemistry was used to determine TF expression and microvessel density. The Student t-test was used to compare high and low TF expression with microvessel density and with age. The chi-squared test was used for other comparisons, and Kaplan-Meier curves were used for survival analyses.
Results Forty-three patients were operated with curative intent. Their mean age was 58.1 ± 12.6 years old, and 62.8% were male. The rectum was the most common location (60,4%), and most tumors reached the serosa and peri-intestinal fat (72.1%). Lymph nodes were positive in 46.5%, and 72.1% of the tumors were moderately differentiated adenocarcinomas. Death occurred in 27.6 ± 12.8 months in 51.1% of the patients who had recurrence. Tissue factor expression was intense in 88.4%. There was a positive correlation between TF expression and microvessel density (p = 0.02), and between TF and older age (p < 0.01). There was no correlation between TF expression and other variables (gender, histological type, penetration into the intestinal wall, and lymphatic and systemic metastases). Tissue factor expression did not correlate with survival.
Conclusion Tissue factor expression correlated with increased microvessel density and older age. Further studies are necessary to ascertain the clinical relevance of TF in colorectal cancer.
Collapse
Affiliation(s)
- Lúcio Sarubbi Fillmann
- PhD in Medicine and Health Sciences from PUC-RS and Professor in the Department of Surgery at PUC-RS School of Medicine, Porto Alegre, RS, Brazil
| | - Laura Pinho Fillmann
- Student of the Undergraduate Course in Medicine at PUC-RS, Porto Alegre, RS, Brazil
| | | | - Henrique Sarubbi Fillmann
- PhD from UFRGS and Professor at the School of Medicine at PUC-RS, Professor at the Department of Surgery at the School of Medicine at PUC-RS, Porto Alegre, RS, Brazil
| | - Gustavo Franco Carvalhal
- PhD in Urology from USP and Professor in the Department of Surgery at the PUC-RS School of Medicine, Porto Alegre, RS, Brazil
| |
Collapse
|
4
|
Gomez S, Tsung A, Hu Z. Current Targets and Bioconjugation Strategies in Photodynamic Diagnosis and Therapy of Cancer. Molecules 2020; 25:E4964. [PMID: 33121022 PMCID: PMC7662882 DOI: 10.3390/molecules25214964] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/18/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
Photodynamic diagnosis (PDD) and therapy (PDT) are emerging, non/minimally invasive techniques for cancer diagnosis and treatment. Both techniques require a photosensitizer and light to visualize or destroy cancer cells. However, a limitation of conventional, non-targeted PDT is poor selectivity, causing side effects. The bioconjugation of a photosensitizer to a tumor-targeting molecule, such as an antibody or a ligand peptide, is a way to improve selectivity. The bioconjugation strategy can generate a tumor-targeting photosensitizer conjugate specific for cancer cells, or ideally, for multiple tumor compartments to improve selectivity and efficacy, such as cancer stem cells and tumor neovasculature within the tumor microenvironment. If successful, such targeted photosensitizer conjugates can also be used for specific visualization and detection of cancer cells and/or tumor angiogenesis (an early event in tumorigenesis) with the hope of an early diagnosis of cancer. The purpose of this review is to summarize some current promising target molecules, e.g., tissue factor (also known as CD142), and the currently used bioconjugation strategies in PDT and PDD, with a focus on newly developed protein photosensitizers. These are genetically engineered photosensitizers, with the possibility of generating a fusion protein photosensitizer by recombinant DNA technology for both PDT and PDD without the need of chemical conjugation. We believe that providing an overview of promising targets and bioconjugation strategies will aid in driving research in this field forward towards more effective, less toxic, and non- or minimally invasive treatment and diagnosis options for cancer patients.
Collapse
Affiliation(s)
- Salvador Gomez
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
- College of Medicine, The Ohio State University, 370 W 9th Ave, Columbus, OH 43210, USA
| | - Allan Tsung
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| | - Zhiwei Hu
- The James-Comprehensive Cancer Center, Division of Surgical Oncology Department of Surgery, College of Medicine, The Ohio State University, 460 W 12th Ave, Columbus, OH 43210, USA; (S.G.); (A.T.)
| |
Collapse
|
5
|
Kawaguchi M, Yamamoto K, Kataoka H, Izumi A, Yamashita F, Kiwaki T, Nishida T, Camerer E, Fukushima T. Protease-activated receptor-2 accelerates intestinal tumor formation through activation of nuclear factor-κB signaling and tumor angiogenesis in Apc Min/+ mice. Cancer Sci 2020; 111:1193-1202. [PMID: 31997435 PMCID: PMC7156842 DOI: 10.1111/cas.14335] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte growth factor activator inhibitor‐1 (HAI‐1), encoded by the SPINT1 gene, is a membrane‐bound protease inhibitor expressed on the surface of epithelial cells. Hepatocyte growth factor activator inhibitor‐1 regulates type II transmembrane serine proteases that activate protease‐activated receptor‐2 (PAR‐2). We previously reported that deletion of Spint1 in ApcMin/+ mice resulted in accelerated formation of intestinal tumors, possibly through enhanced nuclear factor‐κB signaling. In this study, we examined the role of PAR‐2 in accelerating tumor formation in the ApcMin/+ model in the presence or absence of Spint1. We observed that knockout of the F2rl1 gene, encoding PAR‐2, not only eliminated the enhanced formation of intestinal tumors caused by Spint1 deletion, but also reduced tumor formation in the presence of Spint1. Exacerbation of anemia and weight loss associated with HAI‐1 deficiency was also normalized by compound deficiency of PAR‐2. Mechanistically, signaling triggered by deregulated protease activities increased nuclear translocation of RelA/p65, vascular endothelial growth factor expression, and vascular density in ApcMin/+‐induced intestinal tumors. These results suggest that serine proteases promote intestinal carcinogenesis through activation of PAR‐2, and that HAI‐1 plays a critical tumor suppressor role as an inhibitor of matriptase, kallikreins, and other PAR‐2 activating proteases.
Collapse
Affiliation(s)
- Makiko Kawaguchi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Koji Yamamoto
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Aya Izumi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Fumiki Yamashita
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takumi Kiwaki
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takahiro Nishida
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Eric Camerer
- Inserm U970, Paris Cardiovascular Research Center, Université de Paris, Paris, France
| | | |
Collapse
|
6
|
Sherief LM, Hassan TH, Zakaria M, Fathy M, Eshak EA, Bebars MA, Esh A. Tissue factor expression predicts outcome in children with neuroblastoma: A retrospective study. Oncol Lett 2019; 18:6347-6354. [PMID: 31807159 PMCID: PMC6876335 DOI: 10.3892/ol.2019.11021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 05/24/2019] [Indexed: 01/06/2023] Open
Abstract
Previous studies have revealed that the processes of tumor angiogenesis, metastasis and invasiveness are highly dependent on components of the blood coagulation cascade. Tissue factor (TF) is one of the key proteins in coagulation. Cumulative evidence suggested that in addition to its role in coagulation, TF regulates intracellular signaling pathways that serve an important role in angiogenesis, tumor development and metastasis. In the present study, TF expression in neuroblastoma as well as its association with tumor stage, pathology and outcome were assessed. A total of 40 formalin-fixed paraffin-embedded tissues were evaluated for TF expression by immunohistochemical analysis. Results revealed that TF expression was positive in 75% of the analyzed tumor tissues. No significant association between TF expression and sex, age, tumor stage or disease pathology was observed. MYCN proto-oncogene bHLH transcription factor (MYCN) was upregulated in 45% (n=18) of the study cases. Positive TF expression was observed in 94.4% of patients (n=17) with upregulated MYCN, while 59% of patients (n=13) with normal MYCN showed positive TF expression (P<0.05). TF expression was a significant outcome predictor for patients; 18/30 patients (60%) with positive TF expression succumbed to the disease during the study period. In conclusion, TF may be a promising prognosis indicator for neuroblastoma. Future studies to determine how TF affects the progression and outcome of neuroblastoma, as well as to investigate its potential role as a therapeutic target, are required.
Collapse
Affiliation(s)
- Laila M Sherief
- Pediatric Oncology Unit, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| | - Tamer H Hassan
- Pediatric Oncology Unit, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| | - Marwa Zakaria
- Pediatric Oncology Unit, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| | - Manar Fathy
- Pediatric Oncology Unit, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| | - Elia A Eshak
- Department of Pathology, Cairo University, Cairo 11956, Arab Republic of Egypt
| | - Marwa A Bebars
- Pediatric Oncology Unit, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| | - Asmaa Esh
- Department of Clinical Pathology, Zagazig University, Zagazig 44519, Arab Republic of Egypt
| |
Collapse
|
7
|
Challenges in Optimising the Successful Construction of Antibody Drug Conjugates in Cancer Therapy. Antibodies (Basel) 2018; 7:antib7010011. [PMID: 31544863 PMCID: PMC6698866 DOI: 10.3390/antib7010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/21/2022] Open
Abstract
Although considerable progress has been made in the field of cancer chemotherapy, there remains a significant unmet medical need, with a requirement to move away from traditional cytotoxics and explore novel, smarter chemotherapeutic approaches. One such example of the smart chemotherapy approach is antibody-drug conjugates (ADCs), which consist of an antibody that binds selectively to a cancer antigen linked to a cytotoxic agent. When developing an ADC, it may be necessary to produce a variety of constructs to fully assess the optimal configuration for the molecule. By testing ADCs prepared using a range of cytotoxic agents, linkers, or different antibodies, it is possible to fully assess the optimal approach for this treatment modality before advancing to the clinic. Since the development and approval of first-generation ADCs, significant improvements in development technology have occurred. Here, we consider the advances made within the field of ADCs, focusing on the development of EDO-B278 and EDO-B776, both of which have demonstrated efficacy in preclinical testing. Although some limitations remain in this field of development, the potential reduction in toxicity offered by ADCs justifies the investment in research to find workable solutions that could ultimately provide patients with superior outcomes.
Collapse
|
8
|
Wang S, Zhang Y, Cong W, Liu J, Zhang Y, Fan H, Xu Y, Lin H. Breast cancer stem-like cells can promote metastasis by activating
platelets and down-regulating antitumor activity of natural killer
cells. J TRADIT CHIN MED 2017; 36:530-7. [PMID: 28459521 DOI: 10.1016/s0254-6272(16)30071-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To investigate whether cancer stem
cells (CSCs) more efficiently activating platelets and
evading immune surveillance than non-CSCs thus
promoting metastasis. METHODS We enriched and identified sphere-forming
cells (SFCs) and coincubated washed platelets
with several platelet activators including collagen,
4T1 and SFCs. Platelet-coating tumor cells,
platelet activation and TGF-β1 release were analyzed.
Then natural kell cells (NK) were incubated
with supernatants of different activated platelet
samples what we called sample release (SR). The degranulation
assay and NKG2D expression on NK
cells were conducted by flow cytometry. Finally tissue
factor (TF) expression of SFCs or 4T1 were evaluated
by western blot. RESULTS Breast cancer cell line 4T1 could form
spheres in serum-free medium at low adherence.
Sphere-forming cells expressed high levels of the
CD24-/lowCD44 + stem cell phenotype. Both
sphere-forming cells or 4T1 were coated with abundant
platelets while sphere-forming cells induced
significantly higher expression of platelet activating
receptor CD62p than 4T1 did (P < 0.01). And
sphere-forming cells induced platelets to produce
more TGF-β1 than 4T1 did (P < 0.01). Furthermore,
sample releases induced by sphere-forming cells
caused more vigorous inhibition of NK cells antitumor
reactivity (P < 0.05) and reduced NKG2D expression
(P < 0.01). The final results showed that
sphere-forming cells expressed higher levels of TF than 4T1 (P < 0.05). CONCLUSION Our findings indicate that CSCs
could efficiently activate platelets, induce platelets
to secrete more TGF-β1, decrease NKG2D expression
and inhibit antitumor activity of NK cell, compared
with 4T1. And higher levels of TF expression
of CSCs may account for this correlation of CSCs and platelets.
Collapse
|
9
|
Yu G, Li H, Wang X, Wu T, Zhu J, Huang S, Wan Y, Tang J. MicroRNA-19a targets tissue factor to inhibit colon cancer cells migration and invasion. Mol Cell Biochem 2013; 380:239-47. [PMID: 23666757 DOI: 10.1007/s11010-013-1679-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022]
Abstract
The over-expression of tissue factor (TF) and its roles in colon cancer progression have attracted much attention. However, the mechanisms regulating TF expression have not yet been shown in detail. In this study, we over-expressed miR-19a, miR20a and miR-106b in colon cancer cells, and evaluated their impact on TF expression and cellular function. We provide evidence demonstrating that miR-19a inhibited TF expression in vitro. Luciferase reporter assay confirmed that TF was a direct target of miR-19a because the miR-19a mediated repression of luciferase activity was abolished by mutation of the putative binding site. Moreover, miR-19a suppressed colon cancer cell migration and invasion. This effect was due to the indirect down-regulation of matrix metalloproteinase 9. Finally, we investigated the relevance of TF and miR-19a expression in a total of 48 paired colon cancer samples and revealed that miR-19a was inversely correlated with TF expression in stages I and II cases. Therefore, our results suggested that miR-19a was capable of suppressing TF expression in vitro and inhibiting cell migration and invasion. Although it was not the unique mechanism responsible for the expression of TF in vivo, miR-19a was inversely correlated with TF expression in early stage colon cancer patients.
Collapse
Affiliation(s)
- Ge Yu
- Department of General Surgery, First Hospital of Peking University, No. 8 Xishiku Street, West District, Beijing 100034, China
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Yu YJ, Li YM, Hou XD, Guo C, Cao N, Jiao ZY. Effect of tissue factor on invasion inhibition and apoptosis inducing effect of oxaliplatin in human gastric cancer cell. Asian Pac J Cancer Prev 2013; 13:1845-9. [PMID: 22901134 DOI: 10.7314/apjcp.2012.13.5.1845] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Tissue factor (TF) is expressed abnormally in certain types of tumor cells, closely related to invasion and metastasis. The aim of this study was to construct a human gastric cancer cell line SGC7901 stably-transfected with human TF, and observe effects on oxaliplatin-dependent inhibition of invasion and the apoptosis induction. METHODS The target gene TF was obtained from human placenta by nested PCR and introduced into the human gastric cell line SGC7901 through transfection mediated by lipofectamine. Stably-transfected cells were screened using G418. Examples successfully transfected with TF-pcDNA3 recombinant (experimental group), and empty vector pcDNA3 (control group) were incubated with oxaliplatin. Transwell chambers were used to show change in invasive ability. Caspase-3 activity was detected using a colorimetric method and annexin-V/PI double- staining was applied to detect apoptosis. RESULTS We generated the human gastric cancer cell line SGC7901/TF successfully, expressing TF stably and efficiently. Compared with the control group, invasion increased, whereas caspase-3 activity and apoptosis rate were decreased in the experimental group. CONCLUSION TF can enhance the invasive capacity of gastric cancer cells in vitro. Its increased expression may reduce invasion inhibition and apoptosis-inducing effects of oxaliplatin and therefore may warrant targeting for improved chemotherapy.
Collapse
Affiliation(s)
- Yong-Jiang Yu
- Department of General Surgery, First Hospital of Lanzhou Univercity, Lanzhou, China
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
Genetics-based studies have established the critical importance of tumor cell-associated tissue factor, circulating and endothelial cell-associated regulators of thrombin function and multiple thrombin substrates in metastasis. There appear to be multiple pathways by which procoagulants influence tumor biology, but the capacity of hemostatic factors to regulate innate immune function is at least one emerging theme. Several reports have shown that the platelet/fibrin(ogen) axis supports metastasis by limiting natural killer cellmediated lysis of newly-localized micrometastases. Furthermore, there is increasingly compelling evidence that hemostatic and innate immune system interactions also support very early events in cancer development. Analyses of the role of fibrin(ogen) in inflammation-driven colon cancer established a major role for this provisional matrix protein in early tumor development. A seminal property of fibrin(ogen) driving tumor formation in this context is the capacity to support local leukocyte activation events through engagement of the leukocyte integrin α(M)β(2). More recent studies have also suggested that hemostatic factors can, in at least some settings, program the malignant phenotype in tumor cells. Platelet-derived TGF-β1 and other platelet products were reported to trigger a more invasive and prometastatic epithelial-mesenchymal-like transition in embolic tumor cells. These findings support the intriguing concept that tumor cell functional properties can continue to evolve, even beyond the primary tumor site, in response to tumor cell-hemostatic factor interactions in the bloodstream. Taken together, there is strong evidence that the hemostatic system plays a multifaceted role in cancer pathogenesis and that therapies targeting selected hemostatic factors may present a powerful means to impede tumor development and metastasis.
Collapse
Affiliation(s)
- Jay L Degen
- Cancer and Blood Diseases Institute, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | | |
Collapse
|
12
|
Jia ZC, Wan YL, Tang JQ, Dai Y, Liu YC, Wang X, Zhu J. Tissue factor/activated factor VIIa induces matrix metalloproteinase-7 expression through activation of c-Fos via ERK1/2 and p38 MAPK signaling pathways in human colon cancer cell. Int J Colorectal Dis 2012; 27:437-45. [PMID: 22076613 DOI: 10.1007/s00384-011-1351-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2011] [Indexed: 02/04/2023]
Abstract
PURPOSE Increased expression of tissue factor (TF) is associated with tumor invasion and metastasis in human colorectal cancer. We have previously observed that TF/FVIIa upregulates matrix metalloproteinase-7 (MMP-7) expression at the transcriptional level in colon cancer cells. MMP-7 overexpression is believed to play an important role in tumor invasion and metastasis. The aim of this study is to elucidate the molecular mechanisms by which TF/FVIIa induced MMP-7 expression and cell invasion in vitro. METHODS Reverse transcription polymerase chain reaction, Western blot, luciferase assay, and chromatin immunoprecipitation (ChIP) were used to determine the potential mechanism and signaling pathways by which TF/FVIIa induced MMP-7 expression and cell invasion in LoVo cells. Small interfering RNA (siRNA) and cell invasion assay was used to examine whether blocking c-Fos expression could abolish FVIIa-mediated upregulation of MMP-7 and cell invasion in vitro. RESULTS The results showed that FVIIa induced the upregulation of MMP-7 both at the mRNA and protein levels in a time- and dose-dependent manner and increased the invasive behavior of LoVo cells. FVIIa enhanced the promoter activity of MMP-7, and the activator protein-1 (AP-1) binding site was responsible for the activation. Site mutation of the AP-1 binding site in the promoter almost completely abolished FVIIa-mediated response. Furthermore, ChIP assay confirmed that FVIIa promoted the direct binding of c-Fos with the MMP-7 promoter in vivo. FVIIa also induced the expression and nuclear accumulation of the AP-1 subunit c-Fos. siRNA-mediated knockdown of c-Fos eliminated FVIIa-stimulated MMP-7 expression and cell migration in vitro. In addition, selective mitogen-activated protein kinase (MAPK) kinase (MEK1/2) inhibitor (PD98059) and p38 MAPK inhibitor SB203580 suppressed MMP-7 upregulation induced by FVIIa. CONCLUSIONS Our data suggest that a novel TF/FVIIa/MAPK/c-Fos/MMP-7 axis plays an important role in modulating the invasion of colon cancer cells and blockage of this pathway holds promise to treat colon cancer metastasis.
Collapse
Affiliation(s)
- Zhi-Chao Jia
- Laboratory of Colon Cancer, First Hospital of Peking University, No. 8, Xishiku Street, West District, Beijing, 100034, China
| | | | | | | | | | | | | |
Collapse
|
13
|
Ichikawa J, Cole HA, Magnussen RA, Mignemi NA, Butler M, Holt GE, O'Rear L, Yuasa M, Pabla B, Haro H, Cates JMM, Hamm HE, Schwartz HS, Schoenecker JG. Thrombin induces osteosarcoma growth, a function inhibited by low molecular weight heparin in vitro and in vivo. Cancer 2011; 118:2494-506. [DOI: 10.1002/cncr.26518] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/08/2011] [Accepted: 08/03/2011] [Indexed: 12/21/2022]
|
14
|
Lindholm PF, Lu Y, Adley BP, Vladislav T, Jovanovic B, Sivapurapu N, Yang XJ, Kajdacsy-Balla A. Role of monocyte-lineage cells in prostate cancer cell invasion and tissue factor expression. Prostate 2010; 70:1672-82. [PMID: 20607747 DOI: 10.1002/pros.21202] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Tissue factor (TF) is a cell surface glycoprotein intricately related to blood coagulation and inflammation. This study was performed to investigate the role of monocyte-lineage cells in prostate cancer cell TF expression and cell invasion. METHODS Prostate cancer cell invasion was tested with and without added peripheral blood monocytes or human monocyte-lineage cell lines. TF neutralizing antibodies were used to determine the TF requirement for prostate cancer cell invasion activity. Immunohistochemistry was performed to identify prostate tissue CD68 positive monocyte-derived cells and prostate epithelial TF expression. RESULTS Co-culture of PC-3, DU145, and LNCaP cells with isolated human monocytes significantly stimulated prostate cancer cell invasion activity. TF expression was greater in highly invasive prostate cancer cells and was induced in PC-3, DU145, and LNCaP cells by co-culture with U-937 cells, but not with THP-1 cells. TF neutralizing antibodies inhibited PC-3 cell invasion in co-cultures with monocyte-lineage U-937 or THP-1 cells. Prostate cancer tissues contained more CD68 positive cells in the stroma and epithelium (145 ± 53/mm(2)) than benign prostate (108 ± 31/mm(2)). Samples from advanced stage prostate cancer tended to contain more CD68 positive cells when compared with lower stage lesions. Prostatic adenocarcinoma demonstrated significantly increased TF expression compared with benign prostatic epithelium. CONCLUSIONS This study shows that co-culture with monocyte-lineage cells induced prostate cancer cell invasion activity. PC-3 invasion and TF expression was induced in co-culture with U-937 cells and partially inhibited with TF neutralizing antibodies.
Collapse
Affiliation(s)
- Paul F Lindholm
- Department of Pathology, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Zhao J, Aguilar G, Palencia S, Newton E, Abo A. rNAPc2 inhibits colorectal cancer in mice through tissue factor. Clin Cancer Res 2009; 15:208-16. [PMID: 19118048 DOI: 10.1158/1078-0432.ccr-08-0407] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Recombinant nematode anticoagulant protein c2 (rNAPc2) is a specific inhibitor of tissue factor (TF)/factor VIIa complex with novel antithrombotic activity. TF is highly expressed in human colorectal tumors, and levels are positively correlated with disease progression. EXPERIMENTAL DESIGN To explore the therapeutic potential and mechanism of action of rNAPc2 during tumor growth and metastasis, we tested rNAPc2 in several experimental colorectal cancer models in mice. RESULTS Administration of rNAPc2 inhibited pulmonary metastasis in mice systemically disseminated with CT26 murine colon carcinoma cells in a dose-dependent fashion. Combining rNAPc2 with the cytotoxic agent 5-fluorouracil or bevacizumab (humanized anti-vascular endothelial growth factor monoclonal antibody) resulted in additive growth inhibition and simultaneous reduction of microvessel density in HCT116 human colorectal tumor xenografts in nude mice. Furthermore, rNAPc2 potentiated CPT-11 in inhibiting hepatic metastasis in nude mice with portal vein injection of HCT116 tumor cells. Long-term administration of rNAPc2 significantly suppressed spontaneous formation of intestinal tumors in Apc(Min/+) mice. Using a RNA interference approach, we showed that TF expression is necessary for rNAPc2-mediated inhibition of HCT116 human colorectal tumor xenograft growth in nude mice, indicating that the antitumor effect of rNAPc2 may be transduced through TF that is expressed on tumor cells. CONCLUSIONS rNAPc2 is a potent anticancer agent when used in combination with chemotherapy or antiangiogenic therapy in mouse models of colorectal cancer, and TF positivity appears to be required for its activity.
Collapse
Affiliation(s)
- Jingsong Zhao
- Department of Research, Nuvelo, Inc., San Carlos, California 94070, USA.
| | | | | | | | | |
Collapse
|
16
|
Zerbib P, Grimonprez A, Corseaux D, Mouquet F, Nunes B, Petersen LC, Susen S, Ung A, Hebbar M, Pruvot FR, Chambon JP, Jude B. Inhibition of tissue factor-factor VIIa proteolytic activity blunts hepatic metastasis in colorectal cancer. J Surg Res 2008; 153:239-45. [PMID: 19062044 DOI: 10.1016/j.jss.2008.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 04/21/2008] [Accepted: 05/13/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND Expression of the principal initiator of coagulation, tissue factor (TF), by colorectal cancer (CRC) cells is involved in tumoral angiogenesis and metastasis progression, after binding of factor VIIa (FVIIa) to TF and generation of TF-FVIIa activity. We thus hypothesized that inhibition of the TF pathway by active site-blocked FVIIa (FFR-FVIIa) may prevent the development of hepatic metastasis in CRC. METHODS Rat tumoral cells (DHDK12 proB cells) expressing high levels of TF were injected in the portal vein in syngenic BDIX rats. Rats received intraperitoneal injection of either FFR-FVIIa, from d 3 to d 7 (adjuvant treatment) (n = 19), or solvent buffer (n = 18) (control group). Additionally, cancer cells were infused subcutaneously in 20 other rats, which were assigned to FFR-FVIIa adjuvant treatment (n = 10), or buffer treatment (n = 10). Macroscopic and histological analysis was performed at d 14. RESULTS In the control group, infusion of cancer cells resulted in development of macroscopic hepatic tumors in 17/18 rats. In the adjuvant FFR-FVIIa group, macroscopic hepatic tumors were visible on the liver surface in 3/19 rats (P = 0.002 versus control). All rats with subcutaneous injection of proB cells exhibited macroscopic tumors, with no significant difference between the control and the treated ones. CONCLUSION Inhibition of the proteolytic activity of TF-FVIIa complex blunted hematogenous hepatic metastasis, suggesting that TF-FVIIa is a relevant target for the prevention of hepatic metastasis in CRC. TF-blocking agents should be investigated as adjuvant treatment in this setting.
Collapse
Affiliation(s)
- Philippe Zerbib
- Inserm ERI-9, Faculté de Médecine Lille, France; Université de Lille 2, EA2693, Lille, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Palumbo JS, Degen JL. Mechanisms linking tumor cell-associated procoagulant function to tumor metastasis. Thromb Res 2008; 120 Suppl 2:S22-8. [PMID: 18023710 DOI: 10.1016/s0049-3848(07)70127-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Joseph S Palumbo
- Cincinnati Children's Hospital Medical Center, Division of Hematology/Oncology, Cincinnati, OH, USA.
| | | |
Collapse
|
18
|
Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Hu Z, Barney KA, Degen JL. Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood 2007; 110:133-41. [PMID: 17371949 PMCID: PMC1896107 DOI: 10.1182/blood-2007-01-065995] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Tumor cell-associated tissue factor (TF) is a powerful determinant of metastatic potential. TF may increase metastasis by supporting thrombin-mediated proteolysis, through intracellular signaling events mediated by the TF cytoplasmic domain, through TF/fVIIa/fXa-mediated activation of protease-activated receptors, or through a combination of these processes. To better define the relationship between tumor cell-associated TF and circulating hemostatic factors in malignancy, we generated a set of C57Bl/6-derived tumor lines genetically lacking TF, expressing wild-type murine TF, or expressing a mutant TF lacking the cytoplasmic domain. Comparison of the metastatic potential of these cells in immunocompetent mice with genetic deficits in prothrombin, platelet function, or fibrinogen revealed that TF supports metastasis through mechanisms independent of the cytoplasmic domain, but dependent on each of these distal hemostatic factors. TF was neither required for primary tumor growth nor necessary for initial localization of embolized tumor cells within the lungs. Rather, tumor cell fate studies indicated TF supports metastasis by increasing the survival of micrometastases. One mechanism linking TF to metastasis is through a fibrin(ogen)-dependent and platelet-dependent restriction in natural killer cell-mediated clearance of micrometastases. However, TF also supported the early success of micrometastases through an additional mechanism independent of natural killer cells, but coupled to circulating prothrombin.
Collapse
Affiliation(s)
- Joseph S Palumbo
- Divisions of Hematology, Children's Hospital Research Foundation and the University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Haas SL, Jesnowski R, Steiner M, Hummel F, Ringel J, Burstein C, Nizze H, Liebe S, Löhr JM. Expression of tissue factor in pancreatic adenocarcinoma is associated with activation of coagulation. World J Gastroenterol 2006; 12:4843-9. [PMID: 16937466 PMCID: PMC4087618 DOI: 10.3748/wjg.v12.i30.4843] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study expression of tissue factor (TF) in pancreatic cancer and its role in the development of thromboembolism.
METHODS: TF expression was studied in eight human pancreatic carcinoma cell lines by Northern blot and indirect immunofluorescence. Expression of alternatively spliced TF (asTF) was assessed by RT-PCR. In addition, TF expression was determined by immunofluorescence in pancreatic tissues of 19 patients with pancreatic adenocarcinoma (PCa), 9 patients with chronic pancreatitis (CP) and 20 normal controls. Plasma samples (30 PCa-patients, 13 CP-patients and 20 controls) were investigated for soluble TF levels and coagulation activation markers [thrombin-antithrombin III complex (TAT), prothrombin fragment 1 + 2 (F1 + 2)].
RESULTS: All pancreatic carcinoma cell lines expressed TF (8/8) and most of them expressed asTF (6/8). TF expression at the protein level did not correlate with the differentiation of the carcinoma cell line. All but two pancreatic cancer tissue samples stained positive for TF (17/19). In all samples of CP weak staining was restricted to pancreatic duct cells, whereas only a few subendothelial cells were positive in 9/20 of normal controls. TF and TAT levels in PCa patients were significantly elevated compared to controls whereas elevated F1 + 2 levels did not reach statistical significance compared to controls. In CP patients TAT and F1 + 2 levels proved to be significantly elevated compared to controls, although TAT elevation was less pronounced than in PCa patients.
CONCLUSION: We conclude that in addition to the upregulated expression of TF on the cell membrane, soluble TF might contribute to activation of the coagulation system in pancreatic cancer.
Collapse
Affiliation(s)
- Stephan L Haas
- Department of Medicine II, University of Heidelberg at Mannheim, D-68167 Mannheim, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
The large number of conflicting reports on the presence and concentration of circulating tissue factor (TF) in blood generates uncertainties regarding its relevance to hemostasis and association with specific diseases. We believe that the source of these controversies lies in part in the assays used for TF quantitation. We have developed a highly sensitive and specific double monoclonal antibody fluorescence-based immunoassay and integrated it into the Luminex Multi-Analyte Platform. This assay, which uses physiologically relevant standard and appropriate specificity controls, measures TF antigen in recombinant products and natural sources including placenta, plasma, cell lysates and cell membranes. Comparisons of reactivity patterns of various full-length and truncated TFs on an equimolar basis revealed quantitative differences in the immune recognition of TFs by our antibodies in the order of TF 1-263 > 1-242 > 1-218 > placental TF. Despite this differential recognition, all TF species are quantifiable at concentrations < or = 2 pM. Using a calibration curve constructed with recombinant TF 1-263 and plasma from healthy individuals (n = 91), we observed the concentration of TF antigen in plasma to be substantially lower than that generally reported in the literature: TF antigen in plasma of 72 individuals (79%) was below 2 pM (quantitative limit of our assay); TF antigen levels between 2.0 and 5.0 pM could be detected in six individuals (7%); and in 14% (13 plasmas), the non-specific signal was higher than the specific signal, and thus TF levels could not be determined. These differential recognition patterns affect TF quantitation in plasma and should be considered when evaluating plasma TF-like antigen concentrations.
Collapse
Affiliation(s)
- B Parhami-Seren
- Department of Biochemistry, College of Medicine, University of Vermont, Burlington, VT 05446-0068, USA.
| | | | | | | |
Collapse
|
21
|
Abstract
BACKGROUND The improvement of renal allograft survival by pre-transplantation transfusions alerted the medical community to the potential detrimental effect of transfusions in patients being treated for cancer. OBJECTIVES The present meta-analysis aims to evaluate the role of perioperative blood transfusions (PBT) on colorectal cancer recurrence. This is accomplished by validating the results of a previously published meta-analysis (Amato 1998); and by updating it to December 2004. SEARCH STRATEGY Published papers were retrieved using Medline, EMBASE, the Cochrane Library, controlled trials web-based registries, or the CCG Trial Database. The search strategy used was: {colon OR rectal OR colorectal} WITH {cancer OR tumor OR neoplasm} AND transfusion. The tendency not to publish negative trials was balanced by inspecting the proceedings of international congresses. SELECTION CRITERIA Patients undergoing curative resection of colorectal cancer (classified either as Dukes stages A-C, Astler-Coller stages A-C2, or TNM stages T1-3a/N0-1/M0) were included if they had received any amount of blood products within one month of surgery. Excluded were patients with distant metastases at surgery, and studies with short follow-up or with no data. DATA COLLECTION AND ANALYSIS A specific form was developed for data collection. Data extraction was cross-checked, using the most recent publication in case of repetitive ones. Papers' quality was ranked using the method by Evans and Pollock. Odds ratios (OR, with 95% confidence intervals) were computed for each study, and pooled estimates were generated by RevMan (version 4.2). When available, data were stratified for risk factors of cancer recurrence. MAIN RESULTS The findings of the 1998 meta-analysis were confirmed, with small variations in some estimates. Updating it through December 2004 led to the identification of 237 references. Two-hundred and one of them were excluded because they analyzed survival (n=22), were repetitive (n=26), letters/reviews (n=66) or had no data (n=87). Thirty-six studies on 12,127 patients were included: 23 showed a detrimental effect of PBT; 22 used also multivariable analyses, and 14 found PBT to be an independent prognostic factor. Pooled estimates of PBT effect on colorectal cancer recurrence yielded overall OR of 1.42 (95% CI, 1.20 to 1.67) against transfused patients in randomized controlled studies. Stratified meta-analyses confirmed these findings, also when stratifying patients by site and stage of disease. The PBT effect was observed regardless of timing, type, and in a dose-related fashion, although heterogeneity was detected. Data on surgical techniques was not available for further analysis. AUTHORS' CONCLUSIONS This updated meta-analysis confirms the previous findings. All analyses support the hypothesis that PBT have a detrimental effect on the recurrence of curable colorectal cancers. However, since heterogeneity was detected and conclusions on the effect of surgical technique could not be drawn, a causal relationship cannot still be claimed. Carefully restricted indications for PBT seems necessary.
Collapse
Affiliation(s)
- A Amato
- Sigma Tau Research, Inc., 10101 Grosvenor Place, apartment#1415, Rockville, Maryland 20852, USA.
| | | |
Collapse
|
22
|
Han LY, Landen CN, Kamat AA, Lopez A, Bender DP, Mueller P, Schmandt R, Gershenson DM, Sood AK. Preoperative serum tissue factor levels are an independent prognostic factor in patients with ovarian carcinoma. J Clin Oncol 2005; 24:755-61. [PMID: 16380413 DOI: 10.1200/jco.2005.02.9181] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Tissue factor (TF) is a procoagulant that plays an important part in tumor angiogenesis. We sought to determine the role of preoperative serum TF levels in predicting clinical outcome in patients with ovarian cancer. MATERIALS AND METHODS TF expression was determined by reverse transcriptase polymerase chain reaction in ovarian cell lines. Using enzyme-linked immunosorbent assay, we assessed preoperative serum TF levels in 98 women with invasive epithelial ovarian carcinoma, 30 with low malignant potential (LMP) tumors, 16 with benign tumors, and a separate validation group of 39 women with adnexal masses. Clinical information was gathered from chart review. RESULTS TF was expressed in four of the five ovarian cancer cell lines, but absent in the nontransformed cells. Ovarian cancer patients had a median preoperative serum TF level of 85.2 pg/mL, which was significantly higher than in those with LMP tumors (12.8 pg/mL; P < .01) and benign adnexal disease (30.7 pg/mL; P < .01). TF >or= 190 pg/mL was significantly associated with decreased patient survival (P < .01). After adjusting for other clinical variables in a multivariate Cox regression model, TF >or= 190 pg/mL was an independent prognostic factor (P < .01). Analysis of serum TF levels from the validation set confirmed that high TF (>or=190 pg/mL) was associated with a 3.4-fold increase in risk of death from disease (P = .02) and shorter survival (P = .01). CONCLUSION Preoperative serum TF levels are significantly elevated in patients with ovarian carcinoma. Elevated preoperative TF level is an independent prognostic factor for death from disease.
Collapse
Affiliation(s)
- Liz Y Han
- Department of Gynecologic Oncology, The University of Texas M.D. Anderson Cancer Center, Unit 1362, PO Box 301439, Houston, TX 77230-1439, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wang B, Berger M, Masters G, Albone E, Yang Q, Sheedy J, Kirksey Y, Grimm L, Wang B, Singleton J, Soltis D. Radiotherapy of human xenograft NSCLC tumors in nude mice with a 90Y-labeled anti-tissue factor antibody. Cancer Biother Radiopharm 2005; 20:300-9. [PMID: 15989475 DOI: 10.1089/cbr.2005.20.300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Tissue factor (TF) is a type I transmembrane protein and the initiator of the extrinsic blood coagulation pathway. TF plays a critical role in tumor development and its overexpression is observed in many tumors. To understand the prevalence and relative level of TF expression in non-small-cell lung cancer (NSCLC), we analyzed 50 NSCLC tumors by immunohistochemical staining and found that 88% of human NSCLC tumors overexpressed TF. We then generated a high affinity anti-TF antibody, TF278, which specifically binds TF on the surface of cells and is internalized upon binding. An 111In-labeled TF278 demonstrated favorable tumor accumulation in an SW-900 xenograft tumor model with a maximum mean percent of injected dose per gram of tissue (%ID/g) of 73.1% at 96 hours postinjection. In addition, we labeled the antibody with 90Y and tested its ability to inhibit the growth of tumors in an SW-900 xenograft tumor model in immunocompromised mice. The 90Y-TF278 slowed the growth of SW-900 tumors at a 50 microCi dose and completely regressed SW-900 tumors at a 150 microCi dose with little toxicity.
Collapse
Affiliation(s)
- Baiyang Wang
- BioRexis Pharmaceutical Corporation, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The transmembrane glycoprotein tissue factor (TF) is the initiator of the coagulation cascade in vivo. When TF is exposed to blood, it forms a high-affinity complex with the coagulation factors factor VII/activated factor VIIa (FVII/VIIa), activating factor IX and factor X, and ultimately leading to the formation of an insoluble fibrin clot. TF plays an essential role in hemostasis by restraining hemorrhage after vessel wall injury. An overview of biological and physiological aspects of TF, covering aspects consequential for thrombosis and hemostasis such as TF cell biology and biochemistry, blood-borne (circulating) TF, TF associated with microparticles, TF encryption-decryption, and regulation of TF activity and expression is presented. However, the emerging role of TF in the pathogenesis of diseases such as sepsis, atherosclerosis, certain cancers and diseases characterized by pathological fibrin deposition such as disseminated intravascular coagulation and thrombosis, has directed attention to the development of novel inhibitors of tissue factor for use as antithrombotic drugs. The main advantage of inhibitors of the TF*FVIIa pathway is that such inhibitors have the potential of inhibiting the coagulation cascade at its earliest stage. Thus, such therapeutics exert minimal disturbance of systemic hemostasis since they act locally at the site of vascular injury.
Collapse
Affiliation(s)
- Karl-Erik Eilertsen
- Department of Biochemistry, Institute of Medical Biology, Faculty of Medicine, University of Tromsø, Norway.
| | | |
Collapse
|
25
|
Chen C, Yang Q, Patel S, Lei Y, McAleer L, Singleton J, Soltis D, Wang B. Characterization of Human Tissue Factor (TF)–Specific Monoclonal Antibodies Prepared Using a Rapid Immunization Protocol. Hybridoma (Larchmt) 2005; 24:78-85. [PMID: 15857171 DOI: 10.1089/hyb.2005.24.78] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tissue factor (TF) plays an important, physiological role in hemostasis. Recent studies have demonstrated the over-expression of TF in a number of solid tumor types and its pathological roles in angiogenesis and tumor metastasis. In this study, we report the development and characterization of a panel of murine MAbs that are specific for human TF, but do not inhibit TF-mediated blood coagulation. By using a modified repetitive immunizations at multiple sites (RIMMS) protocol in conjunction with an efficient hybridoma cloning procedure, anti-TF MAbs were generated within a relatively short time frame of 5-6 weeks. Following primary screening by ELISA, the binding of the MAbs to the native form of human TF was demonstrated in flow cytometry using a stable cell line expressing human TF. Several of these TF-specific MAbs did not inhibit blood coagulation in a blood coagulation assay and bound with high affinity (0.5-2 nM) to human TF in BIAcore analyses. Importantly, this study represents an independent evaluation of the RIMMS strategy for MAb generation and demonstrates that class-switched, high-affinity MAbs can be generated rapidly and reliably using RIMMS.
Collapse
Affiliation(s)
- Chaoyuan Chen
- Department of Immunotherapeutics, Purdue Pharma L.P., Cranbury, New Jersey, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Stone MD, Harvey SB, Martinez MB, Bach RR, Nelsestuen GL. Large Enhancement of Functional Activity of Active Site-Inhibited Factor VIIa Due to Protein Dimerization: Insights into Mechanism of Assembly/Disassembly from Tissue Factor. Biochemistry 2005; 44:6321-30. [PMID: 15835921 DOI: 10.1021/bi050007z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Active site-inhibited blood clotting factor VIIa (fVIIai) binds to tissue factor (TF), a cell surface receptor that is exposed upon injury and initiates the blood clotting cascade. FVIIai blocks binding of the corresponding enzyme (fVIIa) or zymogen (fVII) forms of factor VII and inhibits coagulation. Although several studies have suggested that fVIIai may have superior anticoagulation effects in vivo, a challenge for use of fVIIai is cost of production. This study reports the properties of dimeric forms of fVIIai that are cross-linked through their active sites. Dimeric wild-type fVIIai was at least 75-fold more effective than monomeric fVIIai in blocking fVIIa association with TF. The dimer of a mutant fVIIai with higher membrane affinity was 1600-fold more effective. Anticoagulation by any form of fVIIai differed substantially from agents such as heparin and showed a delayed mode of action. Coagulation proceeded normally for the first minutes, and inhibition increased as equilibrium binding was established. It is suggested that association of fVIIa(i) with TF in a collision-dependent reaction gives equal access of inhibitor and enzyme to TF. Assembly was not influenced by the higher affinity and slower dissociation of the dimer. As a result, anticoagulation was delayed until the reaction reached equilibrium. Properties of different dissociation experiments suggested that dissociation of fVIIai from TF occurred by a two-step mechanism. The first step was separation of TF-fVIIa(i) while both proteins remained bound to the membrane, and the second step was dissociation of the fVIIa(i) from the membrane. These results suggest novel actions of fVIIai that distinguish it from most of the anticoagulants that block later steps of the coagulation cascade.
Collapse
Affiliation(s)
- Matthew D Stone
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, Minnesota 55455, USA
| | | | | | | | | |
Collapse
|
27
|
Lykke J, Nielsen HJ. Haemostatic alterations in colorectal cancer: perspectives for future treatment. J Surg Oncol 2004; 88:269-75. [PMID: 15565555 DOI: 10.1002/jso.20158] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The role of the haemostatic system in colorectal cancer (CRC) is reviewed. Correlations between the activation of the haemostatic system and overall survival have been suggested. Experimental studies indicate that the haemostatic system plays a key role in growth, invasion and dissemination of tumour cells, and in tumour related angiogenesis. Additional activation by the surgical trauma and postoperative infections are discussed. Finally, anti-cancer modalities directed against regulation of the haemostatic system in CRC are considered.
Collapse
Affiliation(s)
- Jakob Lykke
- Surgical Immunology Laboratory, Department of Surgical Gastroenterology, Hvidovre University Hospital, Copenhagen, Denmark.
| | | |
Collapse
|
28
|
Mälarstig A, Tenno T, Jossan S, Aberg M, Siegbahn A. A quantitative real-time PCR method for tissue factor mRNA. Thromb Res 2004; 112:175-83. [PMID: 14967415 DOI: 10.1016/j.thromres.2003.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2003] [Revised: 10/28/2003] [Accepted: 11/04/2003] [Indexed: 11/23/2022]
Abstract
BACKGROUND Tissue factor (TF) is primarily known for its function to initiate blood coagulation. The range of in vivo expression of TF is wide and requires a dynamic assay for monitoring. A general method for TF mRNA quantitation that is dynamic, sensitive and applicable to a variety of experimental systems or clinical situations is therefore desirable. OBJECTIVES To develop a method for sensitive and dynamic quantitation of TF mRNA in human blood cells. METHODS TF mRNA expression was analysed and evaluated in monocyte isolations, in whole blood (healthy volunteers and patients scheduled for percutaneous coronary intervention, PCI) and in a panel of human cell lines. RNA was extracted, reverse transcribed and subjected to real-time PCR amplification, according to the TaqMan technology. A TF plasmid was constructed as calibrator of the assay. Two housekeeping genes used as endogenous controls for cDNA quality and integrity were evaluated. RESULTS The assay was linear by seven orders of magnitude and detected down to 10(2) copies of the TF plasmid. The coefficient of variation was 4% intra-assay and 28% between the assays when using beta2MG as endogenous control. The beta-actin gene expression was induced by treatment with lipopolysaccharide (LPS) in blood leukocytes and could not be used as an endogenous control. However, beta2MG showed only minor variations upon treatment with LPS. The TF mRNA and antigen expression, measured in a Western blot, correlated well (R(2)=0.903) in a panel of 11 human cell lines. CONCLUSIONS We have established a method for sensitive and dynamic quantitation of TF mRNA in experimental systems and for clinical situations.
Collapse
Affiliation(s)
- Anders Mälarstig
- Department of Medical Sciences, Uppsala University, Uppsala S-75185, Sweden
| | | | | | | | | |
Collapse
|
29
|
|