1
|
Tinholt M, Tekpli X, Torland LA, Tahiri A, Geisler J, Kristensen V, Sandset PM, Iversen N. The breast cancer coagulome in the tumor microenvironment and its role in prognosis and treatment response to chemotherapy. J Thromb Haemost 2024; 22:1319-1335. [PMID: 38237862 DOI: 10.1016/j.jtha.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND The procoagulant phenotype in cancer is linked to thrombosis, cancer progression, and immune response. A novel treatment that reduces the risk of both thrombosis and cancer progression without excess bleeding risk remains to be identified. OBJECTIVES Here, we aimed to broadly investigate the breast tumor coagulome and its relation to prognosis, treatment response to chemotherapy, and the tumor microenvironment. METHODS Key coagulation-related genes (n = 35) were studied in a Norwegian cohort with tumor (n = 134) and normal (n = 189) tissue and in the Cancer Genome Atlas (n = 1052) data set. We performed gene set variation analysis in the Norwegian cohort, and in the Cancer Genome Atlas cohort, associations with the tumor microenvironment and prognosis were evaluated. Analyses were performed with cBioPortal, Estimation of Stromal and Immune cells in Malignant Tumors Using Expression Data, Tumor Immune Estimation Resource, the integrated repository portal for tumor-immune system interactions, Tumor Immune Single-cell Hub 2, and the receiver operating characteristic plotter. Six independent breast cancer cohorts were used to study the tumor coagulome and treatment response to chemotherapy. RESULTS Twenty-two differentially expressed coagulation-related genes were identified in breast tumors. Several coagulome factors were correlated with tumor microenvironment characteristics and were expressed by nonmalignant cells in the tumor microenvironment. PLAT and F8 were independent predictors of better overall survival and progression-free survival, respectively. F12 and PLAU were predictors of worse progression-free survival. The PROCR-THBD-PLAT signature showed a promising predictive value (area under the curve, 0.75; 95% CI, 0.69-0.81; P = 3.6 × 10-17) for combination chemotherapy with fluorouracil, epirubicin, and cyclophosphamide. CONCLUSION The breast tumor coagulome showed potential in prediction of prognosis and chemotherapy response. Cells within the tumor microenvironment are sources of coagulome factors and may serve as therapeutic targets of coagulation factors.
Collapse
Affiliation(s)
- Mari Tinholt
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Department of Haematology, Oslo University Hospital, Oslo, Norway.
| | - Xavier Tekpli
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Lilly Anne Torland
- Department of Research and Innovation, Vestre Viken Hospital Trust, Drammen, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Andliena Tahiri
- Department of Research and Innovation, Vestre Viken Hospital Trust, Drammen, Norway; Department of Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Lørenskog, Norway
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Campus Akershus University Hospital, Lørenskog, Norway
| | - Vessela Kristensen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Per Morten Sandset
- Department of Haematology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Norway
| | - Nina Iversen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Nevskaya KV, Pershina AG, Hmelevskaya ES, Efimova LV, Ibragimova MK, Dolgasheva DS, Tsydenova IA, Ufandeev AA, Buyko EE, Perina EA, Gaptulbarova KA, Kravtsova EA, Krivoshchekov SV, Ivanov VV, Guriev AM, Udut EV, Litviakov NV. Prevention of Metastasis by Suppression of Stemness Genes Using a Combination of microRNAs. J Med Chem 2024; 67:5591-5602. [PMID: 38507819 DOI: 10.1021/acs.jmedchem.3c02199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
We propose an original strategy for metastasis prevention using a combination of three microRNAs that blocks the dedifferentiation of cancer cells in a metastatic niche owing to the downregulation of stemness genes. Transcriptome microarray analysis was applied to identify the effects of a mixture of microRNAs on the pattern of differentially expressed genes in human breast cancer cell lines. Treatment of differentiated CD44- cancer cells with the microRNA mixture inhibited their ability to form mammospheres in vitro. The combination of these three microRNAs encapsulated into lipid nanoparticles prevented lung metastasis in a mouse model of spontaneous metastasis. The mixture of three microRNAs (miR-195-5p/miR-520a/miR-630) holds promise for the development of an antimetastatic therapeutic that blocks tumor cell dedifferentiation, which occurs at secondary tumor sites and determines the transition of micrometastases to macrometastases.
Collapse
Affiliation(s)
- Kseniya V Nevskaya
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Alexandra G Pershina
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
- Research School of Chemical and Biomedical Engineering, Tomsk Polytechnic University, Lenin Ave. 30, Tomsk 634050, Russia
| | - Ekaterina S Hmelevskaya
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Lina V Efimova
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Marina K Ibragimova
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Tomsk State University, Lenin Ave. 36, Tomsk 634050, Russia
| | - Darya S Dolgasheva
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Research School of Chemical and Biomedical Engineering, Tomsk Polytechnic University, Lenin Ave. 30, Tomsk 634050, Russia
| | - Irina A Tsydenova
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Tomsk State University, Lenin Ave. 36, Tomsk 634050, Russia
| | - Alexander A Ufandeev
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Evgeny E Buyko
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Ekaterina A Perina
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Ksenia A Gaptulbarova
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Research School of Chemical and Biomedical Engineering, Tomsk Polytechnic University, Lenin Ave. 30, Tomsk 634050, Russia
| | - Ekaterina A Kravtsova
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Tomsk State University, Lenin Ave. 36, Tomsk 634050, Russia
| | - Sergei V Krivoshchekov
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Vladimir V Ivanov
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Artem M Guriev
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Elena V Udut
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
| | - Nikolai V Litviakov
- Central Research Laboratory, Siberian State Medical University, Moskovsky Trakt 2, Tomsk 634050, Russia
- Oncovirology Lab, Cancer Research Institute of Tomsk National Research Medical Center, Russian Academy of Sciences, Pereulok Kooperativnyi 5, Tomsk 634050, Russia
- Research School of Chemical and Biomedical Engineering, Tomsk Polytechnic University, Lenin Ave. 30, Tomsk 634050, Russia
| |
Collapse
|
3
|
Kroone C, Tieken C, Kocatürk B, Paauwe M, Blok EJ, Ünlü B, van den Berg YW, Stanganello E, Kapteijn MY, Swier N, Zhang X, Duits DEM, Lin Y, Oostenbrink LVE, van den Akker RFP, Mosnier LO, Hawinkels LJ, van Vlijmen BJM, Ruf W, Kuppen PJ, Cannegieter SC, Buijs JT, Versteeg HH. Tumor-expressed factor VII is associated with survival and regulates tumor progression in breast cancer. Blood Adv 2023; 7:2388-2400. [PMID: 36920782 PMCID: PMC10238845 DOI: 10.1182/bloodadvances.2022008455] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Cancer enhances the risk of venous thromboembolism, but a hypercoagulant microenvironment also promotes cancer progression. Although anticoagulants have been suggested as a potential anticancer treatment, clinical studies on the effect of such modalities on cancer progression have not yet been successful for unknown reasons. In normal physiology, complex formation between the subendothelial-expressed tissue factor (TF) and the blood-borne liver-derived factor VII (FVII) results in induction of the extrinsic coagulation cascade and intracellular signaling via protease-activated receptors (PARs). In cancer, TF is overexpressed and linked to poor prognosis. Here, we report that increased levels of FVII are also observed in breast cancer specimens and are associated with tumor progression and metastasis to the liver. In breast cancer cell lines, tumor-expressed FVII drives changes reminiscent of epithelial-to-mesenchymal transition (EMT), tumor cell invasion, and expression of the prometastatic genes, SNAI2 and SOX9. In vivo, tumor-expressed FVII enhanced tumor growth and liver metastasis. Surprisingly, liver-derived FVII appeared to inhibit metastasis. Finally, tumor-expressed FVII-induced prometastatic gene expression independent of TF but required a functional endothelial protein C receptor, whereas recombinant activated FVII acting via the canonical TF:PAR2 pathway inhibited prometastatic gene expression. Here, we propose that tumor-expressed FVII and liver-derived FVII have opposing effects on EMT and metastasis.
Collapse
Affiliation(s)
- Chantal Kroone
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris Tieken
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Begüm Kocatürk
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Madelon Paauwe
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik J. Blok
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Betül Ünlü
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Yascha W. van den Berg
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eliana Stanganello
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Maaike Y. Kapteijn
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Nathalie Swier
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Xi Zhang
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Danique E. M. Duits
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Yazhi Lin
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa V. E. Oostenbrink
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob F. P. van den Akker
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Lukas J. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart J. M. van Vlijmen
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Wolfram Ruf
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Peter J. Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne C. Cannegieter
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen T. Buijs
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H. Versteeg
- Department of Internal Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Ünlü B, Kocatürk B, Rondon AMR, Lewis CS, Swier N, van den Akker RFP, Krijgsman D, Noordhoek I, Blok EJ, Bogdanov VY, Ruf W, Kuppen PJK, Versteeg HH. Integrin regulation by tissue factor promotes cancer stemness and metastatic dissemination in breast cancer. Oncogene 2022; 41:5176-5185. [PMID: 36271029 DOI: 10.1038/s41388-022-02511-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
Tissue Factor (TF) is the initiator of blood coagulation but also functions as a signal transduction receptor. TF expression in breast cancer is associated with higher tumor grade, metastasis and poor survival. The role of TF signaling on the early phases of metastasis has never been addressed. Here, we show an association between TF expression and metastasis as well as cancer stemness in 574 breast cancer patients. In preclinical models, blockade of TF signaling inhibited metastasis tenfold independent of primary tumor growth. TF blockade caused a reduction in epithelial-to-mesenchymal-transition, cancer stemness and expression of the pro-metastatic markers Slug and SOX9 in several breast cancer cell lines and in ex vivo cultured tumor cells. Mechanistically, TF forms a complex with β1-integrin leading to inactivation of β1-integrin. Inhibition of TF signaling induces a shift in TF-binding from α3β1-integrin to α6β4 and dictates FAK recruitment, leading to reduced epithelial-to-mesenchymal-transition and tumor cell differentiation. In conclusion, TF signaling inhibition leads to reduced pro-metastatic transcriptional programs, and a subsequent integrin β1 and β4-dependent reduction in metastasic dissemination.
Collapse
Affiliation(s)
- Betül Ünlü
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Begüm Kocatürk
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Araci M R Rondon
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Clayton S Lewis
- Division of Hematology/Oncology, Department of Internal Medicine, College of Medicine University of Cincinnati, Cincinnati, OH, USA
| | - Nathalie Swier
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob F P van den Akker
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Danielle Krijgsman
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Iris Noordhoek
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik J Blok
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Vladimir Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, College of Medicine University of Cincinnati, Cincinnati, OH, USA
| | - Wolfram Ruf
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
5
|
Regulation of Tissue Factor by CD44 Supports Coagulant Activity in Breast Tumor Cells. Cancers (Basel) 2022; 14:cancers14133288. [PMID: 35805061 PMCID: PMC9266039 DOI: 10.3390/cancers14133288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Metastasis and thromboembolic complications are the main cause of cancer-associated death. An overexpression of coagulation factors, and particularly Tissue factor, by tumor cells is a key event implicated in this observed hypercoagulability. Tissue Factor is indeed a cellular initiator of the coagulation cascade which has been associated with aggressive tumor phenotypes such as those characteristic of Epithelial-Mesenchymal Transitions (EMTs) and Cancer Stem Cells (CSCs). Understanding molecular mechanisms controlling Tissue Factor overexpression in those tumor phenotypes is thus an important aspect of cancer research. We show here that CD44 (a transmembrane marker of CSC and EMT phenotypes) contributes to regulate TF expression at a transcriptional level, thereby supporting procoagulant properties in tumor cells that facilitate their metastatic spread. Abstract Previous work identified Tissue Factor (TF), a key activator of the coagulation cascade, as a gene induced in cellular contexts of Epithelial-Mesenchymal Transitions (EMTs), providing EMT+ Circulating Tumor Cells (CTCs) with coagulant properties that facilitate their metastatic seeding. Deciphering further molecular aspects of TF regulation in tumor cells, we report here that CD44 and TF coexpress in EMT contexts, and that CD44 acts as a regulator of TF expression supporting procoagulant properties and metastatic seeding. A transcriptional regulatory mechanism bridging CD44 to TF expression was further evidenced. Comparing different TF –promoter luciferase reporter constructs, we indeed found that the shortest -111 pb TF promoter fragment harboring three Specificity Protein 1 (Sp1) binding sites is still responsive to CD44 silencing. The observation that (i) mutation within Sp1 binding sites decreased the basal activity of the -111 pb TF promoter construct, (ii) CD44 silencing decreased Sp1 protein and mRNA levels and (iii) Sp1 silencing diminished TF expression further points to Sp1 as a key mediator linking CD44 to TF regulation. All together, these data thus report a transcriptional regulatory mechanism of TF expression by CD44 supporting procoagulant activity and metastatic competence of CTCs.
Collapse
|
6
|
Abstract
Tissue factor (TF), an initiator of extrinsic coagulation pathway, is positively correlated with venous thromboembolism (VTE) of tumor patients. Beyond thrombosis, TF plays a vital role in tumor progression. TF is highly expressed in cancer tissues and circulating tumor cell (CTC), and activates factor VIIa (FVIIa), which increases tumor cells proliferation, angiogenesis, epithelial-mesenchymal transition (EMT) and cancer stem cells(CSCs) activity. Furthermore, TF and TF-positive microvesicles (TF+MVs) activate the coagulation system to promote the clots formation with non-tumor cell components (e.g., platelets, leukocytes, fibrin), which makes tumor cells adhere to clots to form CTC clusters. Then, tumor cells utilize clots to cause its reducing fluid shear stress (FSS), anoikis resistance, immune escape, adhesion, extravasation and colonization. Herein, we review in detail that how TF signaling promotes tumor metastasis, and how TF-targeted therapeutic strategies are being in the preclinical and clinical trials.
Collapse
|
7
|
Wang X, Wan Q, Jin L, Liu C, Liu C, Cheng Y, Wang Z. The Integrative Analysis Identifies Three Cancer Subtypes and Stemness Features in Cutaneous Melanoma. Front Mol Biosci 2021; 7:598725. [PMID: 33665205 PMCID: PMC7921163 DOI: 10.3389/fmolb.2020.598725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/31/2020] [Indexed: 02/03/2023] Open
Abstract
Background: With the growing uncovering of drug resistance in melanoma treatment, personalized cancer therapy and cancer stem cells are potential therapeutic targets for this aggressive skin cancer. Methods: Multi-omics data of cutaneous melanoma were obtained from The Cancer Genome Atlas (TCGA) database. Then, these melanoma patients were classified into different subgroups by performing "CancerSubtypes" method. The differences of stemness indices (mRNAsi and mDNAsi) and tumor microenvironment indices (immune score, stromal score, and tumor purity) among subtypes were investigated. Moreover, the Least Absolute Shrinkage and Selection Operator (LASSO) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) algorithms were performed to identify a cancer cell stemness feature, and the likelihood of immuno/chemotherapeutic response was further explored. Results: Totally, 3 specific subtypes of melanoma with different survival outcomes were identified from TCGA. We found subtype 2 of melanoma with the higher immune score and stromal score and lower mRNAsi and tumor purity score, which has the best survival time than the other subtypes. By performing Kaplan-Meier survival analysis, we found that mRNAsi was significantly associated with the overall survival time of melanomas in subtype 2. Correlation analysis indicated surprising associations between stemness indices and subsets of tumor-infiltrating immune cells. Besides, we developed and validated a prognostic stemness-related genes feature that can divide melanoma patients into high- and low-risk subgroups by applying risk score system. The high-risk group has a significantly shorter survival time than the low-risk subgroup, which is more sensitive to CTLA-4 immune therapy. Finally, 16 compounds were screened out in the Connectivity Map database which may be potential therapeutic drugs for melanomas. Conclusion: Thus, our finding provides a new framework for classification and finds some potential targets for the treatment of melanoma.
Collapse
Affiliation(s)
- Xiaoran Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Qi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Lin Jin
- The First Affiliated Hospital of Shandong First Medical University, Shandong, China
| | - Chengxiu Liu
- Department of Ophthalmology, Affiliated Hospital of Qingdao University Medical College, Qingdao, China
| | - Chang Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yaqi Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Zhichong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
8
|
Hill CN, Hernández-Cáceres MP, Asencio C, Torres B, Solis B, Owen GI. Deciphering the Role of the Coagulation Cascade and Autophagy in Cancer-Related Thrombosis and Metastasis. Front Oncol 2020; 10:605314. [PMID: 33365273 PMCID: PMC7750537 DOI: 10.3389/fonc.2020.605314] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/29/2020] [Indexed: 01/10/2023] Open
Abstract
Thrombotic complications are the second leading cause of death among oncology patients worldwide. Enhanced thrombogenesis has multiple origins and may result from a deregulation of megakaryocyte platelet production in the bone marrow, the synthesis of coagulation factors in the liver, and coagulation factor signaling upon cancer and the tumor microenvironment (TME). While a hypercoagulable state has been attributed to factors such as thrombocytosis, enhanced platelet aggregation and Tissue Factor (TF) expression on cancer cells, further reports have suggested that coagulation factors can enhance metastasis through increased endothelial-cancer cell adhesion and enhanced endothelial cell activation. Autophagy is highly associated with cancer survival as a double-edged sword, as can both inhibit and promote cancer progression. In this review, we shall dissect the crosstalk between the coagulation cascade and autophagic pathway and its possible role in metastasis and cancer-associated thrombosis formation. The signaling of the coagulation cascade through the autophagic pathway within the hematopoietic stem cells, the endothelial cell and the cancer cell are discussed. Relevant to the coagulation cascade, we also examine the role of autophagy-related pathways in cancer treatment. In this review, we aim to bring to light possible new areas of cancer investigation and elucidate strategies for future therapeutic intervention.
Collapse
Affiliation(s)
- Charlotte Nicole Hill
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | | | - Catalina Asencio
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Begoña Torres
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Benjamin Solis
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Walsh CA, Akrap N, Garre E, Magnusson Y, Harrison H, Andersson D, Jonasson E, Rafnsdottir S, Choudhry H, Buffa F, Ragoussis J, Ståhlberg A, Harris A, Landberg G. The mevalonate precursor enzyme HMGCS1 is a novel marker and key mediator of cancer stem cell enrichment in luminal and basal models of breast cancer. PLoS One 2020; 15:e0236187. [PMID: 32692762 PMCID: PMC7373278 DOI: 10.1371/journal.pone.0236187] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
The definitive characterization of common cancer stem cell (CSCs) subpopulations in breast cancer subtypes with distinct genotypic and phenotypic features remains an ongoing challenge. In this study, we have used a non-biased genome wide screening approach to identify transcriptional networks that may be specific to the CSC subpopulations in both luminal and basal breast cancer subtypes. In depth studies of three CSC-enriched breast cancer cell lines representing various subtypes of breast cancer revealed a striking hyperactivation of the mevalonate metabolic pathway in comparison to control cells. The upregulation of metabolic networks is a key feature of tumour cells securing growth and proliferative capabilities and dysregulated mevalonate metabolism has been associated with tumour malignancy and cellular transformation in breast cancer. Furthermore, accumulating evidence suggests that Simvastatin therapy, a mevalonate pathway inhibitor, could affect breast cancer progression and reduce breast cancer recurrence. When detailing the mevalonate pathway in breast cancer using a single-cell qPCR, we identified the mevalonate precursor enzyme, HMGCS1, as a specific marker of CSC-enriched subpopulations within both luminal and basal tumour subtypes. Down-regulation of HMGCS1 also decreased the CSC fraction and function in various model systems, suggesting that HMGCS1 is essential for CSC-activities in breast cancer in general. These data was supported by strong associations between HMGCS1 expression and aggressive features, such as high tumour grade, p53 mutations as well as ER-negativity in lymph node positive breast cancer. Importantly, loss of HMGCS1 also had a much more pronounced effect on CSC-activities compared to treatment with standard doses of Simvastatin. Taken together, this study highlights HMGCS1 as a potential gatekeeper for dysregulated mevalonate metabolism important for CSC-features in both luminal and basal breast cancer subtypes. Pharmacological inhibition of HMGCS1 could therefore be a superior novel treatment approach for breast cancer patients via additional CSC blocking functions.
Collapse
Affiliation(s)
- Claire A. Walsh
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | - Nina Akrap
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | - Elena Garre
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | - Ylva Magnusson
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | - Hannah Harrison
- Paterson Institute for Cancer Research, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Daniel Andersson
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | - Emma Jonasson
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| | | | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Francesca Buffa
- The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Jiannis Ragoussis
- McGill University and Genome Quebec Innovation Centre, Montreal, Canada
| | - Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Adrian Harris
- The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
| | - Göran Landberg
- Sahlgrenska Cancer Center, Department of Laboratory Medicine, Gothenburg, Sweden
| |
Collapse
|
10
|
Unruh D, Horbinski C. Beyond thrombosis: the impact of tissue factor signaling in cancer. J Hematol Oncol 2020; 13:93. [PMID: 32665005 PMCID: PMC7362520 DOI: 10.1186/s13045-020-00932-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Dusten Unruh
- Department of Neurological Surgery, Northwestern University, 303 East Superior St, Chicago, IL, 60611, USA.
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, 303 East Superior St, Chicago, IL, 60611, USA.,Department of Pathology, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
11
|
Guan SS, Wu CT, Liao TZ, Luo TY, Lin KL, Liu SH. Indium-111-labeled CD166-targeted peptide as a potential nuclear imaging agent for detecting colorectal cancer stem-like cells in a xenograft mouse model. EJNMMI Res 2020; 10:13. [PMID: 32096011 PMCID: PMC7040160 DOI: 10.1186/s13550-020-0597-3] [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: 11/25/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are involved in drug resistance, metastasis, and relapse of cancers, which can significantly affect tumor therapy. Hence, to develop specifically therapeutic target probe at CSCs for improvement of survival and quality of life of cancer patients is urgently needed. The CD166 protein has been suggested to be involved in colorectal cancer (CRC) tumorigenesis and to be considered a marker for colorectal CSCs (CRCSCs) detection. In this study, therefore, we attend to apply a nuclear imaging agent probe, Glycine18-Cystine-linked CD166-targeted peptides (CD166tp-G18C), to detect the changes of CD166 level in a CRC xenograft mouse model. RESULTS We isolated the CD166-positive cells from the HCT15 CRC cell line (CD166+HCT15) and evaluated their morphology and ability of clone formation, migration, protein expression, and drug resistance. The CD166-positive HCT15 cells display the CSCs characteristics. We discovered and designed a CD166-targeted peptide (CD166tp-G18C) as a targeted probe of CRC stem-like cell for cell binding assay. The CD166tp-G18C confirmed the CD166 protein targeting ability in CD166+HCT15 cells. The diethylenetriaminopentaacetic acid (DTPA)-conjugated CD166tp-G18C further was labeled with indium-111 (111In-DTPA-CD166tp-G18C) as nuclear imaging agent for imaging and bio-distribution analysis in vivo. Finally, we observed that the 111In-DTPA-CD166tp-G18C was significantly enhanced in tumor tissues of CD166+HCT15 xenograft mice as compared to the non-CD166tp-G18C control. CONCLUSIONS Our results indicated that the indium-111-labeled CD166tp-G18C may be served as a powerful tool for colorectal CSCs nuclear imaging in the CRC patients.
Collapse
Affiliation(s)
- Siao-Syun Guan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Cheng-Tien Wu
- Department of Nutrition, China Medical University, Taichung, 40402, Taiwan.,Master Program of Food and Drug Safety, China Medical University, Taichung, 40402, Taiwan
| | - Tse-Zung Liao
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Tsai-Yueh Luo
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Kun-Liang Lin
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Road, Section 1, Taipei, 10051, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
12
|
Kirwan CC, Descamps T, Castle J. Circulating tumour cells and hypercoagulability: a lethal relationship in metastatic breast cancer. Clin Transl Oncol 2019; 22:870-877. [PMID: 31473984 PMCID: PMC7188731 DOI: 10.1007/s12094-019-02197-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022]
Abstract
Purpose Circulating tumour cells (CTCs) are a marker of poor prognosis and are associated with increased risk of venous thromboembolism in metastatic breast cancer (MBC). We aimed to determine if the presence of CTCs and plasma markers of hypercoagulability [thrombin–antithrombin III (TAT), fibrinogen and d-dimer] are biomarkers of survival in MBC. Methods/patients In a prospective study of MBC patients, CTC (CellSearch®) enumeration and plasma TAT, fibrinogen and d-dimer measured prior to commencement of treatment for disease progression were correlated to overall survival. Results At study completion, of 50 MBC patients recruited (median age 59 years, range 36–82), 40 patients had died (median survival 417 days, range 58–2141). CTCs (≥ 1/7.5 ml) were identified in 16 patients (median number of cells per 7.5 ml, 3 (range 1–31) and were associated with systemic hypercoagulability (medians TAT: 8.1 vs. 5.2 ng/ml, p = 0.03; fibrinogen: 4.3 vs. 3.1 g/l, p = 0.03; d-dimer: 1327 vs. 683 ng/ml, p = 0.0001). At 1 year, of 16 patients with ≥ 1 CTC, 7 had died (44%), compared to 5 of 26 (19%) patients in the no-CTC group. The presence of ≥ 1 CTC was associated with a trend for reduced overall survival (median 455 days vs. 614 days, p = 0.15). Plasma TAT inversely correlated with survival and was significantly higher in patients dying within 1 year (median 9.8 vs. 5.2 ng/ml, p = 0.004) whilst d-dimer showed a trend for reduced 1-year survival (median 1211 vs. 817 ng/ml, p = 0.06). MBC patients with combined high d-dimer (≥ 895 ng/ml) and CTC positivity (≥ 1/7.5 ml whole peripheral blood) had significantly reduced survival (p = 0.04). Conclusions The correlation between CTCs, hypercoagulability and reduced survival in MBC suggests the coagulation system supports tumour cell metastasis and is, therefore, a potential therapeutic target. Electronic supplementary material The online version of this article (10.1007/s12094-019-02197-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- C C Kirwan
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK. .,Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK.
| | - T Descamps
- Centre for Cancer Biomarker Sciences, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield, SK10 4TG, UK
| | - J Castle
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| |
Collapse
|
13
|
Choi C, Thi Thao Tran N, Van Ngu T, Park SW, Song MS, Kim SH, Bae YU, Ayudthaya PDN, Munir J, Kim E, Baek MJ, Song S, Ryu S, Nam KH. Promotion of tumor progression and cancer stemness by MUC15 in thyroid cancer via the GPCR/ERK and integrin-FAK signaling pathways. Oncogenesis 2018; 7:85. [PMID: 30420637 PMCID: PMC6232104 DOI: 10.1038/s41389-018-0094-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/10/2018] [Accepted: 10/03/2018] [Indexed: 12/21/2022] Open
Abstract
Thyroid cancer is the fifth most common cancer diagnosed in women worldwide. Notwithstanding advancements in the prognosis and treatment of thyroid cancer, 10–20% of thyroid cancer patients develops chemotherapeutic resistance and experience relapse. According to previous reports and TCGA database, MUC15 (MUCIN 15) upregulation is highly correlated with thyroid cancer progression. However, the role of MUC15 in tumor progression and metastasis is unclear. This study aimed to investigate factors mediating cancer stemness in thyroid cancer. MUC15 plays an important role in sphere formation, as an evident from the expression of stemness markers including SOX2, KLF4, ALDH1A3, and IL6. Furthermore, ectopic expression of MUC15 activated extracellular signal-regulated kinase (ERK) signaling via G-protein–coupled receptor (GPCR)/cyclic AMP (cAMP) and integrin/focal adhesion kinase pathways. Interestingly, ectopic expression of MUC15 did not affect RAF/mitogen-activated protein kinase kinase (MEK)-mediated ERK activation. The present findings may provide novel insights into the development of diagnostic, prognostic, and therapeutic applications of MUC15 in thyroid cancer.
Collapse
Affiliation(s)
- Cheolwon Choi
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Nguyen Thi Thao Tran
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Trinh Van Ngu
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Sae Woong Park
- Department of Microbiology & Immunology, Weill Cornell Medical College, New York, USA
| | - Min Suk Song
- Department of Life Sciences, Yeungnam University, Gyeongsan, Korea
| | - Sung Hyun Kim
- Department of Physiology, Kyung Hee University, School of Medicine, Seoul, Korea
| | - Yun-Ui Bae
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | | | - Javaria Munir
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Eunbit Kim
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Moo-Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University, Chonan, Korea
| | - Sujung Song
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea
| | - Seongho Ryu
- Soonchunhyang Institute of Med-bioscience (SIMS), Soonchunhyang University, Cheonan, Korea.
| | - Kee-Hyun Nam
- Department of Surgery, College of Medicine, Yonsei University, Seoul, Korea.
| |
Collapse
|
14
|
Graf C, Ruf W. Tissue factor as a mediator of coagulation and signaling in cancer and chronic inflammation. Thromb Res 2018; 164 Suppl 1:S143-S147. [PMID: 29703473 DOI: 10.1016/j.thromres.2018.01.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/23/2022]
Abstract
Thrombosis is frequently diagnosed as a first symptom in tumor patients and the clinical management of hypercoagulability in cancer patients remains challenging due to concomitant changes in risk factors for severe bleeding. It therefore remains a priority to better understand interactions of the hemostatic system with cancer biology. Specifically, further research is needed to elucidate the details and effects of new anticoagulants on extravascular coagulation and the interplay between cancer progression and chronic inflammation. In addition, it will be important to identify subgroups of cancer patients benefiting from specific modulations of the coagulation system without increasing the bleeding risk. Here, we review recent findings on tissue factor (TF) regulation, its procoagulant activity and TF signaling in the various cell types of the tumor microenvironment.
Collapse
Affiliation(s)
- Claudine Graf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany.
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|