1
|
Blümke J, Schameitat M, Verma A, Limbecker C, Arlt E, Kessler SM, Kielstein H, Krug S, Bazwinsky-Wutschke I, Haemmerle M. Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer. Cancers (Basel) 2025; 17:1689. [PMID: 40427186 PMCID: PMC12110028 DOI: 10.3390/cancers17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar-ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Juliane Blümke
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| | - Moritz Schameitat
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Atul Verma
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
| | - Celina Limbecker
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sonja M. Kessler
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Faculty of Natural Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sebastian Krug
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Monika Haemmerle
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| |
Collapse
|
2
|
Chou SC, Chang SL, Yu CY, Lin CI, Chang YT, Chen LF, Li JY, Pai CH, Lin SR, Cheng WC, Yuan CT, Lin SW. Tissue factor, factor VIII and IX in microvesicle-induced thrombosis and tumor growth of pancreatic cancer. Thromb J 2025; 23:32. [PMID: 40217494 PMCID: PMC11987238 DOI: 10.1186/s12959-025-00715-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Tissue factor (TF)-rich cancer microvesicles are correlated with thrombosis risk. Intrinsic coagulation factors are also associated with the risk of thrombosis in cancer patients. This study explored the roles of pancreatic cancer-derived microvesicles and intrinsic factors in thrombogenesis. METHODS Human pancreatic cancer cell lines rich in TF (AsPC-1-TFhigh, MIAPaCa-2-TFhigh) or poor in TF [AsPC-1-TFKO(knockout) and MIAPaCa-2-TFlow] were generated for microvesicle preparation and injected into coagulation-defective mice. Inferior vena cava (IVC) clots and lung thrombosis were evaluated. Immunodeficient hemophilia A (NSG-HA) mice were orthotopically injected with the cells mentioned above, and the tumor and IVC clot weights were analyzed. RESULTS With the injection of TFhigh microvesicles, IVC clots were rarely found in hemophilic mice. The TFlow and TFKO microvesicles resulted in few IVC clots in any mouse. Lung thrombosis was substantially reduced in the hemophilic mice infused with any microvesicle type. In orthotopic tumor models, TFhigh cells grew faster than did TFlow cells. TFhigh tumor-bearing NSG-WT mice had the most enormous IVC clots, whereas NSG-HA mice had no IVC clots. CONCLUSION Pancreatic cancer thrombosis induced by TF-expressing microvesicles strongly depended on FVIII and FIX, while VWF played a minor role. Moreover, TF, but not FVIII, was significantly related to tumor growth.
Collapse
Affiliation(s)
- Sheng-Chieh Chou
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Lun Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Cheng-Yeh Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chao-I Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Yen-Ting Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Li-Fu Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Jia-Yi Li
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chen-Hsueh Pai
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shu-Rung Lin
- Department of Bioscience Technology, College of Science, Chung-Yuan Christian University, Taoyuan, Taiwan
- Center for Nanotechnology, Chung-Yuan Christian University, Taoyuan, Taiwan
| | - Wern-Cherng Cheng
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chang-Tsu Yuan
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, College of Medicine, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Tadokoro H, Ota Y, Uomoto M, Koizume S, Sato S, Nakamura Y, Yoshihara M, Endo-Takahashi Y, Negishi Y, Miyagi E, Miyagi Y. A mouse model of deep vein thrombosis by inferior vena cava hypoperfusion using ameroid constrictors. Sci Rep 2025; 15:928. [PMID: 39762377 PMCID: PMC11704261 DOI: 10.1038/s41598-024-84443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Traditional mouse models for deep vein thrombosis (DVT), frequently utilized in research focused on cancer-associated thrombosis (CAT), reliably induce thrombus formation by obstructing blood flow (BF) in the inferior vena cava (IVC), which does not occur in humans. Therefore, to develop a new DVT model for CAT studies, we implanted an ameroid constrictor (AC), a hygroscopic casein C-shape device, around the IVC and aorta of immunocompromised mice. We evaluated the thrombus 3 and 8 days post-AC implantation and compared it with the traditional model 2 days post-vena cava ligation. The size of each thrombus was measured, and the composition was assessed using histological staining; BF through the IVC was confirmed using ultrasound imaging. The thrombus size variability in the AC and ligation models was equivalent. Compared with thrombi on day 3 post-AC implantation, those on day 8 showed characteristics of human thrombi in the subacute to chronic stage. The BF in the IVC was maintained even on day 8. In summary, the AC model showed reproducibility with no significant difference in thrombus size variability from the traditional ligation model while maintaining the BF of the IVC.
Collapse
Affiliation(s)
- Hiroko Tadokoro
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, 2-3-2 Nakao, Asahi-ku, Yokohama, 241- 8515, Kanagawa, Japan
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, 192-0392, Tokyo, Japan
| | - Yukihide Ota
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Kanagawa, Japan
| | - Mari Uomoto
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Kanagawa, Japan
| | - Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, 2-3-2 Nakao, Asahi-ku, Yokohama, 241- 8515, Kanagawa, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Department of Pathology, Kanagawa Cancer Center Hospital, 2-3-2 Nakao, Asahi-ku, Yokohama, 241- 8515, Kanagawa, Japan
- Morphological Information Analysis Laboratory, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241‑8515, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan
- Morphological Information Analysis Laboratory, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241‑8515, Japan
| | - Mitsuyo Yoshihara
- Morphological Information Analysis Laboratory, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241‑8515, Japan
- Division of Advanced Cancer Therapeutics, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, , Yokohama, 241-8515, Kanagawa, Japan
| | - Yoko Endo-Takahashi
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, 192-0392, Tokyo, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, 192-0392, Tokyo, Japan
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Kanagawa, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, 241-8515, Kanagawa, Japan.
- Department of Pathology, Kanagawa Cancer Center Hospital, 2-3-2 Nakao, Asahi-ku, Yokohama, 241- 8515, Kanagawa, Japan.
| |
Collapse
|
4
|
Ünlü B, Heestermans M, Laghmani EH, Buijs JT, van den Akker RFP, van Vlijmen BJM, Versteeg HH. The effects of an aggressive breast tumor on thrombosis after antithrombin downregulation in a hypercoagulable mouse model. Thromb Res 2024; 244:109200. [PMID: 39476730 DOI: 10.1016/j.thromres.2024.109200] [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: 02/04/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Despite improvements in therapy, breast cancer still contributes to high mortality rates. Survival of these patients becomes progressively worse upon diagnosis with cancer-associated thrombosis (CAT). Unfortunately, the mechanism causing CAT has remained unclear. OBJECTIVE Set up an acute and non-invasive hypercoagulable mouse model with an aggressive breast cancer and study the mechanism of cancer-associated thrombosis. METHODS Mice were grafted with the aggressive breast cancer cell line MDA-MB-231 or sham-treated. Subsequently, an acute imbalance in coagulation was introduced by injecting a synthetic small interfering (si) RNA targeting hepatic Serpinc1 to knockdown antithrombin - a condition known to predispose to cause a hypercoagulant state in vivo. RESULTS Silencing Serpinc1 with siRNA decreased plasma antithrombin levels. siRNA treatment had no short-term effects on tumor characteristics, but increased distant metastasis within the timeframe of this study. The systemic pro-inflammatory status, with elevated platelet counts and fibrinogen levels in tumor-bearing mice, was also not affected by antithrombin silencing. While elevated fibrin deposition in the liver upon Serpinc1 targeting was not significantly affected by the presence of breast cancer, knockdown of antithrombin did significantly increase intratumoral fibrin deposition and inflammation. Surprisingly, in the presence of an aggressive tumor, a protective outcome with less clinical features coinciding with venous thrombosis were observed in mice with antithrombin knockdown. CONCLUSION We conclude that the presence of a breast tumor protects hypercoagulant mice from severe consumption of coagulation factors after lowering hepatic antithrombin levels, possibly due to elevated platelet counts. However, the consequences on cancer-associated thrombosis remained inconclusive.
Collapse
Affiliation(s)
- Betül Ünlü
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Marco Heestermans
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - El Houari Laghmani
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob F P van den Akker
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart J M van Vlijmen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
5
|
Mathews R, Setthavongsack N, Le-Cook A, Kaempf A, Loftis JM, Woltjer RL, Lorentz CU, Revenko A, Hinds MT, Nguyen KP. Role of platelet count in a murine stasis model of deep vein thrombosis. Platelets 2024; 35:2290916. [PMID: 38099327 PMCID: PMC10805383 DOI: 10.1080/09537104.2023.2290916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/06/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023]
Abstract
Platelets are core components of thrombi but their effect on thrombus burden during deep vein thrombosis (DVT) has not been fully characterized. We examined the role of thrombopoietin-altered platelet count on thrombus burden in a murine stasis model of DVT. To modulate platelet count compared to baseline, CD1 mice were pretreated with thrombopoietin antisense oligonucleotide (THPO-ASO, 56% decrease), thrombopoietin mimetic (TPO-mimetic, 36% increase), or saline (within 1%). Thrombi and vein walls were examined on postoperative days (POD) 3 and 7. Thrombus weights on POD 3 were not different between treatment groups (p = .84). The mean thrombus weights on POD 7 were significantly increased in the TPO-mimetic cohort compared to the THPO-ASO (p = .005) and the saline (p = .012) cohorts. Histological grading at POD 3 revealed a significantly increased smooth muscle cell presence in the thrombi and CD31 positive channeling in the vein wall of the TPO-mimetic cohort compared to the saline and THPO-ASO cohorts (p < .05). No differences were observed in histology on POD 7. Thrombopoietin-induced increased platelet count increased thrombus weight on POD 7 indicating platelet count may regulate thrombus burden during early resolution of venous thrombi in this murine stasis model of DVT.
Collapse
Affiliation(s)
- Rick Mathews
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Naly Setthavongsack
- Division of Neuropathology, Department of Pathology, Oregon Health and Science University, Portland, Oregon, USA
| | - Anh Le-Cook
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
| | - Andy Kaempf
- Biostatistics Shared Resource, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Jennifer M Loftis
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
- Department of Psychiatry, Oregon Health and Science University, Portland, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Randall L Woltjer
- Division of Neuropathology, Department of Pathology, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | - Monica T Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
| | - Khanh P Nguyen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon, USA
- Research & Development Service, VA Portland Health Care System, Portland, Oregon, USA
- Division of Vascular Surgery, Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Kawano T, Englisch C, Hisada Y, Paul D, Archibald S, Grover S, Pabinger I, Ay C, Mackman N. Mucin 1 and venous thrombosis in tumor-bearing mice and patients with cancer. Thromb Res 2024; 237:23-30. [PMID: 38547690 PMCID: PMC11058007 DOI: 10.1016/j.thromres.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION Mucins released from epithelial tumors have been proposed to play a role in cancer-associated thrombosis. Mucin1 (MUC1) is a transmembrane mucin that is overexpressed in a variety of human malignancies, including breast and pancreatic cancer. We analyzed the association of MUC1 and venous thrombosis in a mouse tumor model and in patients with cancer. MATERIALS AND METHODS We used a human pancreatic cancer cell line HPAF-II that expresses a high level of MUC1. We grew HPAF-II tumors in the pancreas of Crl:NU-Foxn1nu male mice. MUC1 in plasma and extracellular vesicles (EVs) isolated from plasma was measured using an enzyme-linked immunosorbent assay. MUC1 in EVs and venous thrombi from tumor-bearing mice was assessed by western blotting. We measured MUC1 in plasma from healthy controls and patients with stomach, colorectal or pancreatic cancer with or without venous thromboembolism. RESULTS AND DISCUSSION MUC1 was detected in the plasma of mice bearing HPAF-II tumors and was associated with EVs. MUC1 was present in venous thrombi from mice bearing HFAP-II tumors. Recombinant MUC1 did not induce platelet aggregation. Levels of MUC1 were higher in patients with pancreatic cancer compared with healthy controls. In contrast to the mouse model, MUC1 was present in EV-free plasma in samples from healthy controls and patients with cancer. There was no significant difference in the levels of MUC1 in cancer patients with or without VTE. Our data did not find any evidence that MUC1 contributed to VTE in patients with cancer.
Collapse
Affiliation(s)
- Tomohiro Kawano
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cornelia Englisch
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Paul
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sierra Archibald
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven Grover
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ingrid Pabinger
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Su K, Duan R, Wu Y. Prognostic value of venous thromboembolism in patients with advanced pancreatic cancer: a systematic review and meta-analysis. Front Oncol 2024; 14:1331706. [PMID: 38390258 PMCID: PMC10882063 DOI: 10.3389/fonc.2024.1331706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
OBJECTIVE This study aimed to investigate the relationship between the incidence of VTE and the prognosis of patients with advanced pancreatic cancer, as there is currently a lack of systematic research on this topic, despite the prevalence of venous thromboembolism (VTE) in patients with pancreatic cancer. METHODS Databases including PubMed, Embase, Web of Science, and Cochrane Library were searched until April 9, 2023, to identify studies that explored the relationship between VTE and the prognosis of advanced pancreatic cancer. Duplicate publications, studies without full text or sufficient information for data extraction, animal experiments, reviews, and systematic reviews were excluded. The extracted data were analyzed using STATA 15.1. RESULTS The pooled results indicated a significant association between the incidence of VTE and poorer overall survival (HR=1.38, 95% CI: 1.24 - 1.53, p < 0.001) and disease-free survival (HR=2.42, 95% CI: 1.94 - 3.04, p < 0.001) among patients with advanced pancreatic cancer. Additionally, early VTE showed a significant impact on overall survival (HR=2.03, 95% CI: 1.33 - 3.12, p = 0.001), whereas late VTE did not demonstrate a significant association with poor overall survival (HR=1.22, 95% CI: 0.96 - 1.54, p = 0.099). CONCLUSIONS This study found that advanced pancreatic cancer patients with VTE had poorer overall and disease-free survival than those without. Meanwhile, the patients with early VTE had a significantly poorer prognosis, whereas late VTE did not. The findings highlight the importance of timely detection of VTE for patients with advanced pancreatic cancer patients and offer a partial theoretical basis for future clinical endeavors. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023427043, identifier CRD42023427043.
Collapse
Affiliation(s)
- Kaifeng Su
- Medical Faculty of Ludwig‐Maximilians‐University of Munich, University Hospital of LMU Munich, Munich, Germany
| | - Ruifeng Duan
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Xu Y, Nipper MH, Dominguez AA, Ye Z, Akanuma N, Lopez K, Deng JJ, Arenas D, Sanchez A, Sharkey FE, Court CM, Singhi AD, Wang H, Fernandez-Zapico ME, Sun LZ, Zheng S, Chen Y, Liu J, Wang P. Reconstitution of human PDAC using primary cells reveals oncogenic transcriptomic features at tumor onset. Nat Commun 2024; 15:818. [PMID: 38280869 PMCID: PMC10821902 DOI: 10.1038/s41467-024-45097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024] Open
Abstract
Animal studies have demonstrated the ability of pancreatic acinar cells to transform into pancreatic ductal adenocarcinoma (PDAC). However, the tumorigenic potential of human pancreatic acinar cells remains under debate. To address this gap in knowledge, we expand sorted human acinar cells as 3D organoids and genetically modify them through introduction of common PDAC mutations. The acinar organoids undergo dramatic transcriptional alterations but maintain a recognizable DNA methylation signature. The transcriptomes of acinar organoids are similar to those of disease-specific cell populations. Oncogenic KRAS alone do not transform acinar organoids. However, acinar organoids can form PDAC in vivo after acquiring the four most common driver mutations of this disease. Similarly, sorted ductal cells carrying these genetic mutations can also form PDAC, thus experimentally proving that PDACs can originate from both human acinar and ductal cells. RNA-seq analysis reveal the transcriptional shift from normal acinar cells towards PDACs with enhanced proliferation, metabolic rewiring, down-regulation of MHC molecules, and alterations in the coagulation and complement cascade. By comparing PDAC-like cells with normal pancreas and PDAC samples, we identify a group of genes with elevated expression during early transformation which represent potential early diagnostic biomarkers.
Collapse
Affiliation(s)
- Yi Xu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Michael H Nipper
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Angel A Dominguez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Zhenqing Ye
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Naoki Akanuma
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Kevin Lopez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Janice J Deng
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Destiny Arenas
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Ava Sanchez
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Francis E Sharkey
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Colin M Court
- Division of Surgical Oncology and Endocrine Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Huamin Wang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lu-Zhe Sun
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jun Liu
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| | - Pei Wang
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
9
|
Mackman N, Tawil N, Rak J. Tissue factor at the crossroads of coagulation and radiation response in glioblastoma. J Thromb Haemost 2024; 22:3-6. [PMID: 38173243 DOI: 10.1016/j.jtha.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 01/05/2024]
Affiliation(s)
- Nigel Mackman
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, North Carolina, USA; University of North Carolina at Chapel Hill Blood Research Center, The University of North Carolina at Chapel Hill, North Carolina, USA.
| | - Nadim Tawil
- Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Janusz Rak
- Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Su Y, Yi J, Zhang Y, Leng D, Huang X, Shi X, Zhang Y. EML4-ALK fusion protein in Lung cancer cells enhances venous thrombogenicity through the pERK1/2-AP-1-tissue factor axis. J Thromb Thrombolysis 2024; 57:67-81. [PMID: 37940761 PMCID: PMC10830642 DOI: 10.1007/s11239-023-02916-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Accumulating evidence links the echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) rearrangement to venous thromboembolism (VTE) in non-small cell lung cancer (NSCLC) patients. However, the corresponding mechanisms remain unclear. METHOD High-throughput sequencing analysis of H3122 human ALK-positive NSCLC cells treated with ALK inhibitor/ dimethyl sulfoxide (DMSO) was performed to identify coagulation-associated differential genes between EML4-ALK fusion protein inhibited cells and control cells. Sequentially, we confirmed its expression in NSCLC patients' tissues and in the plasma of a subcutaneous xenograft mouse model. An inferior vena cava (IVC) ligation model was used to assess clot formation potential. Additionally, pathways involved in tissue factor (TF) regulation were explored in ALK-positive cell lines H3122 and H2228. Statistical significance was determined by Student t-test and one-way ANOVA using SPSS. RESULTS Sequencing analysis identified a significant downregulation of TF after inhibiting EML4-ALK fusion protein activity in H3122 cells. In clinical NSCLC cases, TF expression was increased especially in ALK-positive NSCLC tissues. Meanwhile, H3122 and H2228 with high TF expression exhibited shorter plasma clotting time and higher TF activity versus ALK-negative H1299 and A549 in cell culture supernatant. Mice bearing H2228 tumor showed a higher concentration of tumor-derived TF and TF activity in plasma and the highest adjusted IVC clot weights. Limiting EML4-ALK protein phosphorylation downregulated extracellular regulated protein kinases 1/2 (ERK1/2)-activating the protein-1(AP-1) signaling pathway and thus attenuated TF expression. CONCLUSION EML4-ALK fusion protein may enhance venous thrombogenicity by regulating coagulation factor TF expression. There was potential involvement of the pERK1/2-AP-1 pathway in this process.
Collapse
Affiliation(s)
- Yanping Su
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiawen Yi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Dong Leng
- Clinical Laboratory, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xiaoxi Huang
- Basic Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Xinyu Shi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
11
|
Hisada Y, Kawano T, Archibald SJ, Welch JS, Reeves BN, Mackman N. Tissue factor activates the coagulation cascade in mouse models of acute promyelocytic leukemia. Blood Adv 2023; 7:5458-5469. [PMID: 37450381 PMCID: PMC10515313 DOI: 10.1182/bloodadvances.2023010466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023] Open
Abstract
Acute promyelocytic leukemia (APL) is associated with a high risk of bleeding and thrombosis. APL patients have an activated coagulation system, hyperfibrinolysis, and thrombocytopenia. APL cells express tissue factor (TF), a receptor and cofactor for factor VII/VIIa. This study had 2 goals. Firstly, we measured biomarkers of coagulation and fibrinolysis activation as well as platelet counts and bleeding in both mouse xenograft and allograft models of APL. Secondly, we determined the effect of inhibiting TF on the activation of coagulation in these models. We observed increased levels of plasma thrombin-antithrombin complexes (TAT), D-dimer, and plasmin-antiplasmin complexes, reduced platelet counts, and increased tail bleeding in both mouse models of APL. Fibrinogen levels decreased in the xenograft model but not in the allograft model. In contrast, the red blood cell count decreased in the allograft model but not in the xenograft model. Inhibition of APL-derived human TF with an anti-human TF monoclonal antibody reduced the level of TAT, increased platelet count, and normalized tail bleeding in a xenograft model. Inhibition of all sources of TF (APL cells and host cells) in the allograft model with a rat anti-mouse TF monoclonal antibody decreased the levels of TAT but did not affect the platelet count. Our study demonstrates that TF plays a central role in the activation of coagulation in both the xenograft and allograft mouse models of APL. These APL mouse models can be used to investigate the mechanisms of coagulopathy and thrombocytopenia in APL.
Collapse
Affiliation(s)
- Yohei Hisada
- University of North Carolina Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tomohiro Kawano
- University of North Carolina Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Sierra J. Archibald
- University of North Carolina Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - John S. Welch
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO
| | - Brandi N. Reeves
- University of North Carolina Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- University of North Carolina Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
12
|
Wahab R, Hasan MM, Azam Z, Grippo PJ, Al-Hilal TA. The role of coagulome in the tumor immune microenvironment. Adv Drug Deliv Rev 2023; 200:115027. [PMID: 37517779 PMCID: PMC11099942 DOI: 10.1016/j.addr.2023.115027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
The rising incidence and persistent thrombosis in multiple cancers including those that are immunosuppressive highlight the need for understanding the tumor coagulome system and its role beyond hemostatic complications. Immunotherapy has shown significant benefits in solid organ tumors but has been disappointing in the treatment of hypercoagulable cancers, such as glioblastoma and pancreatic ductal adenocarcinomas. Thus, targeting thrombosis to prevent immunosuppression seems a clinically viable approach in cancer treatment. Hypercoagulable tumors often develop fibrin clots within the tumor microenvironment (TME) that dictates the biophysical characteristics of the tumor tissue. The application of systems biology and single-cell approaches highlight the potential role of coagulome or thrombocytosis in shaping the tumor immune microenvironment (TIME). In-depth knowledge of the tumor coagulome would provide unprecedented opportunities to better predict the hemostatic complications, explore how thrombotic stroma modulates tumor immunity, reexamine the significance of clinical biomarkers, and enable steering the stromal versus systemic immune response for boosting the effectiveness of immune checkpoint inhibitors in cancer treatment. We focus on the role of coagulation factors in priming a suppressive TIME and the huge potential of existing anticoagulant drugs in the clinical settings of cancer immunotherapy.
Collapse
Affiliation(s)
- Riajul Wahab
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Md Mahedi Hasan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Zulfikar Azam
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79968, USA; Department of Environmental Science & Engineering, College of Science, University of Texas at El Paso, El Paso, TX 79968, USA.
| |
Collapse
|
13
|
Okeke EB, Louttit C, Snyder CM, Moon JJ. Neutrophils and neutrophil extracellular traps in cancer: promising targets for engineered nanomaterials. Drug Deliv Transl Res 2023; 13:1882-1895. [PMID: 36182992 PMCID: PMC10066838 DOI: 10.1007/s13346-022-01243-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2022] [Indexed: 01/07/2023]
Abstract
Neutrophils are the most abundant white blood cells in circulation and constitute up to 60% of circulating leukocytes. Neutrophils play a significant role in host defense against pathogens through various mechanisms, including phagocytosis, production of antimicrobial proteins, and formation of neutrophil extracellular traps (NETs). Recently, the role of neutrophils and NETs in cancer has generated significant interest, as accumulating evidence suggests that neutrophils and NETs contribute to cancer progression and are associated with adverse patient outcomes. In this review, we will first highlight the roles of neutrophils and NETs in cancer progression and metastasis and discuss new drug delivery approaches to target and modulate neutrophils and NETs for cancer therapeutics.
Collapse
Affiliation(s)
- Emeka B Okeke
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, 14063, USA.
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Cameron Louttit
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Caitlin M Snyder
- Department of Biology, State University of New York at Fredonia, Fredonia, NY, 14063, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
14
|
Kawano T, Hisada Y, Grover SP, Schug WJ, Paul DS, Bergmeier W, Mackman N. Decreased Platelet Reactivity and Function in a Mouse Model of Human Pancreatic Cancer. Thromb Haemost 2023; 123:501-509. [PMID: 36716775 PMCID: PMC10820933 DOI: 10.1055/s-0043-1761419] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cancer patients have increased thrombosis and bleeding compared with the general population. Cancer is associated with activation of both platelets and coagulation. Mouse models have been used to study the dysregulation of platelets and coagulation in cancer. We established a mouse model of pancreatic cancer in which tissue factor-expressing human pancreatic tumors (BxPC-3) are grown in nude mice. Tumor-bearing mice have an activated coagulation system and increased venous thrombosis compared to control mice. We also showed that tumor-derived, tissue factor-positive extracellular vesicles activated platelets ex vivo and in vivo. In this study, we determined the effect of tumors on a platelet-dependent arterial thrombosis model. Unexpectedly, we observed significantly reduced carotid artery thrombosis in tumor-bearing mice compared to controls. In addition, we observed significantly increased tail bleeding in tumor-bearing mice compared to controls. These results suggested that the presence of the tumor affected platelets. Indeed, tumor-bearing mice exhibited a significant decrease in platelet count and an increase in mean platelet volume and percentage of reticulated platelets, findings that are consistent with increased platelet turnover. Levels of the platelet activation marker platelet factor 4 were also increased in tumor-bearing mice. We also observed decreased platelet receptor expression in tumor-bearing mice and reduced levels of active αIIb/β3 integrin in response to PAR4 agonist peptide and convulxin in platelets from tumor-bearing mice compared with platelets from control mice. In summary, our study suggests that in tumor-bearing mice there is chronic platelet activation, leading to thrombocytopenia, decreased receptor expression, and impaired platelet adhesive function.
Collapse
Affiliation(s)
- Tomohiro Kawano
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Yohei Hisada
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Steven P. Grover
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Wyatt J. Schug
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - David S. Paul
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
15
|
Gi (魏 峻洸) T, Kuwahara (桑原 彩) A, Yamashita (山下 篤) A, Matsuda (松田 俊太郎) S, Maekawa (前川 和也) K, Moriguchi-Goto (盛口 淸香) S, Sato (佐藤 勇一郎) Y, Asada (浅田 祐士郎) Y. Histopathological Features of Cancer-Associated Venous Thromboembolism: Presence of Intrathrombus Cancer Cells and Prothrombotic Factors. Arterioscler Thromb Vasc Biol 2023; 43:146-159. [PMID: 36384269 PMCID: PMC9762717 DOI: 10.1161/atvbaha.122.318463] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Cancer-associated venous thromboembolism (VTE) is a critical complication in patients with cancer. However, the pathological findings of VTE are limited. Here, we investigated the histopathological features of cancer-associated VTE in human autopsy cases. METHODS We clinically examined the autopsy cases of VTE with (n=114) and without cancer (n=66) and immunohistochemically analyzed the expression of prothrombotic factors in intrathrombus cancer cells, the thrombus contents of erythrocytes, fibrin, platelets, citrullinated histone H3, and degree of organization. RESULTS Vascular wall invasion or small cell clusters of cancer cells was observed in thrombi in 27.5% of deep vein thrombosis and 25.9% of pulmonary embolism cases. The majority of the cancer cells in deep vein thrombi appeared to be invading the vessel wall, whereas the majority of pulmonary thrombi had cancer cell clusters, consistent with embolization via blood flow. These cancer cells were immunohistochemically positive for TF (tissue factors) or podoplanin in up to 88% of VTE cases. The frequency of TF-positive monocyte/macrophages in thrombi was higher in cancer-associated VTE than that in VTE without cancer. Citrullinated histone H3 was predominantly observed in the early stages of organizing thrombi. There was no significant difference in thrombus components between VTE with cancer and without cancer groups. CONCLUSIONS Vascular wall invasion or cancer cell clusters in thrombi might influence thrombogenesis of cancer-associated VTE. TF and podoplanin in cancer cells and in monocyte/macrophages may induce coagulation reactions and platelet aggregation. Neutrophil extracellular traps may play a role in the early stages of VTE, regardless of cancer status.
Collapse
Affiliation(s)
- Toshihiro Gi (魏 峻洸)
- Department of Pathology (T.G., A.Y., K.M., Y.A.), Faculty of Medicine, University of Miyazaki, Japan
| | - Aya Kuwahara (桑原 彩)
- Department of Laboratory Center (A.K.), Faculty of Medicine, University of Miyazaki, Japan
| | - Atsushi Yamashita (山下 篤)
- Department of Pathology (T.G., A.Y., K.M., Y.A.), Faculty of Medicine, University of Miyazaki, Japan
| | - Shuntaro Matsuda (松田 俊太郎)
- Department of Medicine and Community Health (S.M.), Faculty of Medicine, University of Miyazaki, Japan
| | - Kazunari Maekawa (前川 和也)
- Department of Pathology (T.G., A.Y., K.M., Y.A.), Faculty of Medicine, University of Miyazaki, Japan
| | - Sayaka Moriguchi-Goto (盛口 淸香)
- Department of Diagnostic Pathology‚ University of Miyazaki Hospital (S.M.-G., Y.S.), Faculty of Medicine, University of Miyazaki, Japan
| | - Yuichiro Sato (佐藤 勇一郎)
- Department of Diagnostic Pathology‚ University of Miyazaki Hospital (S.M.-G., Y.S.), Faculty of Medicine, University of Miyazaki, Japan
| | - Yujiro Asada (浅田 祐士郎)
- Department of Pathology (T.G., A.Y., K.M., Y.A.), Faculty of Medicine, University of Miyazaki, Japan
| |
Collapse
|
16
|
Tissue factor in cancer-associated thromboembolism: possible mechanisms and clinical applications. Br J Cancer 2022; 127:2099-2107. [PMID: 36097177 PMCID: PMC9467428 DOI: 10.1038/s41416-022-01968-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/29/2023] Open
Abstract
Venous and arterial thromboses, called as cancer-associated thromboembolism (CAT), are common complications in cancer patients that are associated with high mortality. The cell-surface glycoprotein tissue factor (TF) initiates the extrinsic blood coagulation cascade. TF is overexpressed in cancer cells and is a component of extracellular vesicles (EVs). Shedding of TF+EVs from cancer cells followed by association with coagulation factor VII (fVII) can trigger the blood coagulation cascade, followed by cancer-associated venous thromboembolism in some cancer types. Secretion of TF is controlled by multiple mechanisms of TF+EV biogenesis. The procoagulant function of TF is regulated via its conformational change. Thus, multiple steps participate in the elevation of plasma procoagulant activity. Whether cancer cell-derived TF is maximally active in the blood is unclear. Numerous mechanisms other than TF+EVs have been proposed as possible causes of CAT. In this review, we focused on a wide variety of regulatory and shedding mechanisms for TF, including the effect of SARS-CoV-2, to provide a broad overview for its role in CAT. Furthermore, we present the current technical issues in studying the relationship between CAT and TF.
Collapse
|
17
|
Moik F, Ay C. Hemostasis and cancer: Impact of haemostatic biomarkers for the prediction of clinical outcomes in patients with cancer. J Thromb Haemost 2022; 20:2733-2745. [PMID: 36106749 PMCID: PMC9827869 DOI: 10.1111/jth.15880] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 01/13/2023]
Abstract
Patients with cancer are characterized by a dysregulation of the hemostatic system and systemic hypercoagulability. Different components of the hemostatic system are involved in tumor-promoting mechanisms including primary tumor growth, cancer cell invasion, immune evasion, angiogenesis, and the metastatic process. Therefore, different degrees of systemic hemostatic activation in patients with cancer can reflect distinct underlying biological phenotypes of cancer and seem to correlate with cancer aggressiveness. Peripheral blood levels of hemostatic biomarkers, indicating the activation status of different parts of the hemostatic system including the coagulation cascade, fibrinolytic activity, platelet activation, or endothelial activation, can be used to reflect cancer-associated systemic hypercoagulability. Thereby, hemostatic biomarkers represent promising candidates to investigate as surrogate markers for underlying cancer activity and progression dynamics and therefore as biomarkers for the prediction of clinical outcomes in cancer patients. In the present review, we provide an up-to-date summary of available data on hemostatic biomarkers for prognostication of overall survival and prediction of therapy response in patients with cancer, including specific oncologic treatment settings for potential clinical application. We provide a thorough discussion on potential clinical implementation and current limitations and highlight the most promising emerging biomarkers that might be used to contribute to risk-stratified, personalized oncologic decision making in the future.
Collapse
Affiliation(s)
- Florian Moik
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of ViennaViennaAustria
- Clinical Division of Oncology, Department of Internal Medicine, Medical University of GrazGrazAustria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of ViennaViennaAustria
| |
Collapse
|
18
|
Enhanced Permeability and Retention Effect as a Ubiquitous and Epoch-Making Phenomenon for the Selective Drug Targeting of Solid Tumors. J Pers Med 2022; 12:jpm12121964. [PMID: 36556185 PMCID: PMC9784116 DOI: 10.3390/jpm12121964] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
In 1979, development of the first polymer drug SMANCS [styrene-co-maleic acid (SMA) copolymer conjugated to neocarzinostatin (NCS)] by Maeda and colleagues was a breakthrough in the cancer field. When SMANCS was administered to mice, drug accumulation in tumors was markedly increased compared with accumulation of the parental drug NCS. This momentous result led to discovery of the enhanced permeability and retention effect (EPR effect) in 1986. Later, the EPR effect became known worldwide, especially in nanomedicine, and is still believed to be a universal mechanism for tumor-selective accumulation of nanomedicines. Some research groups recently characterized the EPR effect as a controversial concept and stated that it has not been fully demonstrated in clinical settings, but this erroneous belief is due to non-standard drug design and use of inappropriate tumor models in investigations. Many research groups recently provided solid evidence of the EPR effect in human cancers (e.g., renal and breast), with significant diversity and heterogeneity in various patients. In this review, we focus on the dynamics of the EPR effect and restoring tumor blood flow by using EPR effect enhancers. We also discuss new applications of EPR-based nanomedicine in boron neutron capture therapy and photodynamic therapy for solid tumors.
Collapse
|
19
|
Chen Y, Liu J, Su Y, Zhao H, Zhao Y, Wen M, Lu S, Cao X, Zhang W, Liu L, Wu J. Annexin V - and tissue factor + microparticles as biomarkers for predicting deep vein thrombosis in patients after joint arthroplasty. Clin Chim Acta 2022; 536:169-179. [PMID: 36191610 DOI: 10.1016/j.cca.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Venous thromboembolism (VTE) is a common and severe complication of joint arthroplasty. Microparticles (MPs) containing phosphatidylserine (PS) and tissue factor (TF) can trigger coagulation in VTE. This study aims to measure and compare MP levels in joint arthroplasty patients with and without VTE. METHODS This prospective cohort study enrolled 181 patients who underwent joint arthroplasty. Ultrasound examination was used to diagnose VTE on preoperative day 0 and postoperative day 6. MPs were analysed using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and flow cytometry. The levels of platelet-derived microparticles (PMPs), endothelial cell-derived microparticles (EMPs), granulocyte-derived microparticles (GMPs), red cell-derived microparticles (RMPs), monocyte-derived microparticles (MMPs), Annexin V+ MPs (AV+ MPs), and tissue factor+ MPs (TF+ MPs) derived from five kinds of MPs were measured on day 0 (before surgery), 1, 2, 3, 4, 5, and 6 after surgery. RESULTS The levels of AV-TF+ EMPs and AV-TF+ MMPs were significantly increased in patients with VTE on postoperative day 5 compared to those without VTE (P=0.031 and P=0.031, respectively). CONCLUSION AV-TF+ MPs may indicate the development of VTE and serve as predictive markers in joint arthroplasty patients.
Collapse
Affiliation(s)
- Yuying Chen
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Jian Liu
- Adult reconstruction department, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yu Su
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Huiru Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Yujing Zhao
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Meng Wen
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Shan Lu
- Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China
| | - Xiangyu Cao
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Wenjie Zhang
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China
| | - Lei Liu
- Department of Clinical Laboratory, Liyuan Hospital of Tongji Medical College of Huazhong University of Science and Technology, Hubei, P.R.China
| | - Jun Wu
- Department of Clinical Laboratory, Peking University Fourth School of Clinical Medicine, Beijing, P.R.China; Department of Clinical Laboratory, Beijing Jishuitan Hospital, Beijing, P.R.China.
| |
Collapse
|
20
|
Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J Clin Med 2022; 11:jcm11164932. [PMID: 36013171 PMCID: PMC9410115 DOI: 10.3390/jcm11164932] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan E. El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-01-350-000 (ext. 4765)
| |
Collapse
|
21
|
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
|
22
|
Hisada Y, Sachetto ATA, Mackman N. Circulating tissue factor-positive extracellular vesicles and their association with thrombosis in different diseases. Immunol Rev 2022; 312:61-75. [PMID: 35708588 DOI: 10.1111/imr.13106] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022]
Abstract
Tissue factor (TF) is a procoagulant protein released from activated host cells, such as monocytes, and tumor cells on extracellular vesicles (EVs). TF + EVs are observed in the circulation of patients with various types of diseases. In this review, we will summarize the association between TF + EVs and activation of coagulation and survival in different types of diseases, including cancer, sepsis, and infections with different viruses, such as human immunodeficiency virus (HIV), influenza A virus (IAV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We will also discuss the source of TF + EVs in various diseases. EVTF activity is associated with thrombosis in pancreatic cancer patients and coronavirus disease 2019 patients (COVID-19) and with disseminated intravascular coagulation in cancer patients. EVTF activity is also associated with worse survival in patients with cancer and COVID-19. Monocytes are the major sources of TF + EVs in sepsis, and viral infections, such as HIV, Ebola virus, and SARS-CoV-2. In contrast, alveolar epithelial cells are the major source of TF + EVs in bronchoalveolar lavage fluid in COVID-19 and influenza A patients. These studies indicate that EVTF activity could be used as a biomarker to identify patients that have an increased risk of coagulopathy and mortality.
Collapse
Affiliation(s)
- Yohei Hisada
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ana Teresa Azevedo Sachetto
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Chen Z, Wei X, Dong S, Han F, He R, Zhou W. Challenges and Opportunities Associated With Platelets in Pancreatic Cancer. Front Oncol 2022; 12:850485. [PMID: 35494001 PMCID: PMC9039220 DOI: 10.3389/fonc.2022.850485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/15/2022] [Indexed: 01/02/2023] Open
Abstract
Pancreatic cancer is one of the most common malignant tumors in the digestive system with a poor prognosis. Accordingly, better understanding of the molecular mechanisms and innovative therapies are warranted to improve the prognosis of this patient population. In addition to playing a crucial role in coagulation, platelets reportedly contribute to the growth, invasion and metastasis of various tumors, including pancreatic cancer. This narrative review brings together currently available evidence on the impact of platelets on pancreatic cancer, including the platelet-related molecular mechanisms of cancer promotion, pancreatic cancer fibrosis, immune evasion, drug resistance mechanisms, thrombosis, targeted platelet therapy, combined radiotherapy and chemotherapy treatment, platelet combined with nanotechnology treatment and potential applications of pancreatic cancer organoids. A refined understanding of the role of platelets in pancreatic cancer provides the foothold for identifying new therapeutic targets.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaodong Wei
- Emergency Department, Gansu Provincial Hospital, Lanzhou, China
| | - Shi Dong
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Fangfang Han
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ru He
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
24
|
Abstract
Cancer-associated thrombosis (including venous thromboembolism (VTE) and arterial events) is highly consequential for patients with cancer and is associated with worsened survival. Despite substantial improvements in cancer treatment, the risk of VTE has increased in recent years; VTE rates additionally depend on the type of cancer (with pancreas, stomach and primary brain tumours having the highest risk) as well as on individual patient's and cancer treatment factors. Multiple cancer-specific mechanisms of VTE have been identified and can be classified as mechanisms in which the tumour expresses proteins that alter host systems, such as levels of platelets and leukocytes, and in which the tumour expresses procoagulant proteins released into the circulation that directly activate the coagulation cascade or platelets, such as tissue factor and podoplanin, respectively. As signs and symptoms of VTE may be non-specific, diagnosis requires clinical assessment, evaluation of pre-test probability, and objective diagnostic testing with ultrasonography or CT. Risk assessment tools have been validated to identify patients at risk of VTE. Primary prevention of VTE (thromboprophylaxis) has long been recommended in the inpatient and post-surgical settings, and is now an option in the outpatient setting for individuals with high-risk cancer. Anticoagulant therapy is the cornerstone of therapy, with low molecular weight heparin or newer options such as direct oral anticoagulants. Personalized treatment incorporating risk of bleeding and patient preferences is essential, especially as a diagnosis of VTE is often considered by patients even more distressing than their cancer diagnosis, and can severely affect the quality of life. Future research should focus on current knowledge gaps including optimizing risk assessment tools, biomarker discovery, next-generation anticoagulant development and implementation science.
Collapse
|
25
|
Gauchel N, Krauel K, Hamad MA, Bode C, Duerschmied D. Thromboinflammation as a Driver of Venous Thromboembolism. Hamostaseologie 2021; 41:428-432. [PMID: 34942655 DOI: 10.1055/a-1661-0257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Thrombus formation has been identified as an integral part in innate immunity, termed immunothrombosis. Activation of host defense systems is known to result in a procoagulant environment. In this system, cellular players as well as soluble mediators interact with each other and their dysregulation can lead to the pathological process of thromboinflammation. These mechanisms have been under intensified investigation during the COVID-19 pandemic. In this review, we focus on the underlying mechanisms leading to thromboinflammation as one trigger of venous thromboembolism.
Collapse
Affiliation(s)
- Nadine Gauchel
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Krystin Krauel
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Muataz Ali Hamad
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Duerschmied
- Department of Cardiology and Angiology I, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
26
|
Mai S, Inkielewicz-Stepniak I. Pancreatic Cancer and Platelets Crosstalk: A Potential Biomarker and Target. Front Cell Dev Biol 2021; 9:749689. [PMID: 34858977 PMCID: PMC8631477 DOI: 10.3389/fcell.2021.749689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Platelets have been recognized as key players in hemostasis, thrombosis, and cancer. Preclinical and clinical researches evidenced that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between cancer cells and platelets. Pancreatic cancer is a devastating disease with high morbidity and mortality worldwide. Although the relationship between pancreatic cancer and platelets in clinical diagnosis is described, the interplay between pancreatic cancer and platelets, the underlying pathological mechanism and pathways remain a matter of intensive study. This review summaries recent researches in connections between platelets and pancreatic cancer. The existing data showed different underlying mechanisms were involved in their complex crosstalk. Typically, pancreatic tumor accelerates platelet aggregation which forms thrombosis. Furthermore, extracellular vesicles released by platelets promote communication in a neoplastic microenvironment and illustrate how these interactions drive disease progression. We also discuss the advantages of novel model organoids in pancreatic cancer research. A more in-depth understanding of tumor and platelets crosstalk which is based on organoids and translational therapies may provide potential diagnostic and therapeutic strategies for pancreatic cancer progression.
Collapse
Affiliation(s)
- Shaoshan Mai
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
27
|
Cao W, Zhu MY, Lee SH, Lee SB, Kim HJ, Park BO, Yoon CH, Khadka D, Oh GS, Shim H, Kwak TH, So HS. Modulation of Cellular NAD + Attenuates Cancer-Associated Hypercoagulability and Thrombosis via the Inhibition of Tissue Factor and Formation of Neutrophil Extracellular Traps. Int J Mol Sci 2021; 22:ijms222112085. [PMID: 34769515 PMCID: PMC8584923 DOI: 10.3390/ijms222112085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer-associated thrombosis is the second-leading cause of mortality in patients with cancer and presents a poor prognosis, with a lack of effective treatment strategies. NAD(P)H quinone oxidoreductase 1 (NQO1) increases the cellular nicotinamide adenine dinucleotide (NAD+) levels by accelerating the oxidation of NADH to NAD+, thus playing important roles in cellular homeostasis, energy metabolism, and inflammatory responses. Using a murine orthotopic 4T1 breast cancer model, in which multiple thrombi are generated in the lungs at the late stage of cancer development, we investigated the effects of regulating the cellular NAD+ levels on cancer-associated thrombosis. In this study, we show that dunnione (a strong substrate of NQO1) attenuates the prothrombotic state and lung thrombosis in tumor-bearing mice by inhibiting the expression of tissue factor and formation of neutrophil extracellular traps (NETs). Dunnione increases the cellular NAD+ levels in lung tissues of tumor-bearing mice to restore the declining sirtuin 1 (SIRT1) activity, thus deacetylating nuclear factor-kappa B (NF-κB) and preventing the overexpression of tissue factor in bronchial epithelial and vascular endothelial cells. In addition, we demonstrated that dunnione abolishes the ability of neutrophils to generate NETs by suppressing histone acetylation and NADPH oxidase (NOX) activity. Overall, our results reveal that the regulation of cellular NAD+ levels by pharmacological agents may inhibit pulmonary embolism in tumor-bearing mice, which may potentially be used as a viable therapeutic approach for the treatment of cancer-associated thrombosis.
Collapse
Affiliation(s)
- Wa Cao
- Center for Metabolic Function Regulation and Department of Microbiology, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (W.C.); (M.-Y.Z.); (S.-H.L.); (S.-B.L.)
| | - Meng-Yu Zhu
- Center for Metabolic Function Regulation and Department of Microbiology, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (W.C.); (M.-Y.Z.); (S.-H.L.); (S.-B.L.)
| | - Seung-Hoon Lee
- Center for Metabolic Function Regulation and Department of Microbiology, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (W.C.); (M.-Y.Z.); (S.-H.L.); (S.-B.L.)
| | - Su-Bin Lee
- Center for Metabolic Function Regulation and Department of Microbiology, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (W.C.); (M.-Y.Z.); (S.-H.L.); (S.-B.L.)
| | - Hyung-Jin Kim
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Byung-Ouk Park
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Cheol-Hwan Yoon
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Dipendra Khadka
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Gi-Su Oh
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Hyeok Shim
- Internal Medicine, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea;
| | - Tae-Hwan Kwak
- NADIANBIO Ltd., R201-1, Business Incubation Center, 460 Iksan-daero, Iksan 54538, Jeonbuk, Korea; (H.-J.K.); (B.-O.P.); (C.-H.Y.); (D.K.); (G.-S.O.); (T.-H.K.)
| | - Hong-Seob So
- Center for Metabolic Function Regulation and Department of Microbiology, School of Medicine, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (W.C.); (M.-Y.Z.); (S.-H.L.); (S.-B.L.)
- Correspondence:
| |
Collapse
|
28
|
Wang J, Wang X, Guo Y, Ye L, Li D, Hu A, Cai S, Yuan B, Jin S, Zhou Y, Li Q, Zheng L, Tong Q. Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancer cells and neutrophils inhibits aerobic glycolysis and cancer progression. Clin Transl Med 2021; 11:e588. [PMID: 34841706 PMCID: PMC8567044 DOI: 10.1002/ctm2.588] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND As a metabolic reprogramming feature, cancer cells derive most of their energy from aerobic glycolysis, while its regulatory mechanisms and therapeutic strategies continue to be illusive. METHODS Integrative analysis of publically available expression profile datasets was used to identify critical transcriptional regulators and their target glycolytic enzymes. The functions and acting mechanisms of transcriptional regulators in cancer cells were investigated by using in vitro and in vivo assays. The Kaplan-Meier curve and log-rank assay were used to conduct the survival study. RESULTS Salmonella pathogenicity island 1 (SPI1/PU.1), a haematopoietic transcription factor, was identified to facilitate glycolytic process, tumourigenesis, invasiveness, as well as metastasis of colon cancer cells, which was interplayed by tumour-associated neutrophils. Mechanistically, neutrophils delivered SPI1 mRNA via extracellular vesicles, resulting in enhanced SPI1 expression of cancer cells. Through physical interaction with SPI1-related protein (SPIB), SPI1 drove expression of glycolytic genes within cancer cells, which in turn induced polarization of neutrophils via glycolytic metabolite lactate. Depletion of neutrophils or SPIB-SPI1 interaction in cancer cells significantly inhibited glycolytic process, tumourigenesis and aggressiveness. Upregulation of SPI1 or SPIB was found to be associated with poor prognosis in patients suffering from colon cancer. CONCLUSIONS Therapeutic targeting of SPIB/SPI1-facilitated interplay of cancerous cells and neutrophils suppresses aerobic glycolysis and progression of cancer.
Collapse
Affiliation(s)
- Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Xiaojing Wang
- Department of Geriatrics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yanhua Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Lin Ye
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Anpei Hu
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shuang Cai
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Boling Yuan
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Shikai Jin
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Yi Zhou
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
- Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei Province430022P. R. China
| |
Collapse
|
29
|
Moik F, Prager G, Thaler J, Posch F, Wiedemann S, Schramm T, Englisch C, Mackman N, Pabinger I, Ay C. Hemostatic Biomarkers and Venous Thromboembolism Are Associated With Mortality and Response to Chemotherapy in Patients With Pancreatic Cancer. Arterioscler Thromb Vasc Biol 2021; 41:2837-2847. [PMID: 34470475 DOI: 10.1161/atvbaha.121.316463] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Florian Moik
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Gerald Prager
- Clinical Division of Oncology (G.P.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Johannes Thaler
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Florian Posch
- Division of Haematology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Austria (F.P.)
| | - Sarah Wiedemann
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Theresa Schramm
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Cornelia Englisch
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill (N.M.)
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology (F.M., J.T., S.W., T.S., C.E., I.P., C.A.), Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria.,I. M. Sechenov First Moscow State Medical University, Russia (C.A.)
| |
Collapse
|
30
|
Zifkos K, Dubois C, Schäfer K. Extracellular Vesicles and Thrombosis: Update on the Clinical and Experimental Evidence. Int J Mol Sci 2021; 22:ijms22179317. [PMID: 34502228 PMCID: PMC8431093 DOI: 10.3390/ijms22179317] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Extracellular vesicles (EVs) compose a heterogenous group of membrane-derived particles, including exosomes, microvesicles and apoptotic bodies, which are released into the extracellular environment in response to proinflammatory or proapoptotic stimuli. From earlier studies suggesting that EV shedding constitutes a cellular clearance mechanism, it has become evident that EV formation, secretion and uptake represent important mechanisms of intercellular communication and exchange of a wide variety of molecules, with relevance in both physiological and pathological situations. The putative role of EVs in hemostasis and thrombosis is supported by clinical and experimental studies unraveling how these cell-derived structures affect clot formation (and resolution). From those studies, it has become clear that the prothrombotic effects of EVs are not restricted to the exposure of tissue factor (TF) and phosphatidylserines (PS), but also involve multiplication of procoagulant surfaces, cross-linking of different cellular players at the site of injury and transfer of activation signals to other cell types. Here, we summarize the existing and novel clinical and experimental evidence on the role and function of EVs during arterial and venous thrombus formation and how they may be used as biomarkers as well as therapeutic vectors.
Collapse
Affiliation(s)
- Konstantinos Zifkos
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, D-55131 Mainz, Germany;
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, Institut National de la Recherche pour l’Agriculture, l’alimentation et l’Environnement (INRAE) 1260, Center for CardioVascular and Nutrition Research (C2VN), F-13380 Marseille, France;
| | - Katrin Schäfer
- Department of Cardiology, Cardiology I, University Medical Center Mainz, D-55131 Mainz, Germany
- Correspondence:
| |
Collapse
|
31
|
Hisada Y, Mackman N. Tissue Factor and Extracellular Vesicles: Activation of Coagulation and Impact on Survival in Cancer. Cancers (Basel) 2021; 13:cancers13153839. [PMID: 34359742 PMCID: PMC8345123 DOI: 10.3390/cancers13153839] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The tissue factor (TF)-factor VIIa complex is the major physiological initiator of blood coagulation. Tumors express TF and release TF-positive extracellular vesicles (EVs) into the circulation, and this is associated with the activation of coagulation. Circulating levels of EVTF activity may be a useful biomarker to identify patients at risk for thrombosis. Tumor TF and TF-positive EVs are also associated with reduced survival. Abstract Tissue factor (TF) is a transmembrane glycoprotein that functions as a receptor for FVII/FVIIa and initiates the extrinsic coagulation pathway. Tumors and cancer cells express TF that can be released in the form of TF positive (TF+) extracellular vesicles (EVs). In this review, we summarize the studies of tumor TF and TF + EVs, and their association with activation of coagulation and survival in cancer patients. We also summarize the role of tumor-derived TF + EVs in venous thrombosis in mouse models. Levels of tumor TF and TF + EVs are associated with venous thromboembolism in pancreatic cancer patients. In addition, levels of EVTF activity are associated with disseminated intravascular coagulation in cancer patients. Furthermore, tumor-derived TF + EVs enhance venous thrombosis in mice. Tumor TF and TF + EVs are also associated with worse survival in cancer patients, particularly in pancreatic cancer patients. These studies indicate that EVTF activity could be used as a biomarker to identify pancreatic cancer patients at risk for venous thrombosis and cancer patients at risk for disseminated intravascular coagulation. EVTF activity may also be a useful prognostic biomarker in cancer patients.
Collapse
|
32
|
Burgos-Ravanal R, Campos A, Díaz-Vesga MC, González MF, León D, Lobos-González L, Leyton L, Kogan MJ, Quest AFG. Extracellular Vesicles as Mediators of Cancer Disease and as Nanosystems in Theranostic Applications. Cancers (Basel) 2021; 13:3324. [PMID: 34283059 PMCID: PMC8268753 DOI: 10.3390/cancers13133324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer remains a leading cause of death worldwide despite decades of intense efforts to understand the molecular underpinnings of the disease. To date, much of the focus in research has been on the cancer cells themselves and how they acquire specific traits during disease development and progression. However, these cells are known to secrete large numbers of extracellular vesicles (EVs), which are now becoming recognized as key players in cancer. EVs contain a large number of different molecules, including but not limited to proteins, mRNAs, and miRNAs, and they are actively secreted by many different cell types. In the last two decades, a considerable body of evidence has become available indicating that EVs play a very active role in cell communication. Cancer cells are heterogeneous, and recent evidence reveals that cancer cell-derived EV cargos can change the behavior of target cells. For instance, more aggressive cancer cells can transfer their "traits" to less aggressive cancer cells and convert them into more malignant tumor cells or, alternatively, eliminate those cells in a process referred to as "cell competition". This review discusses how EVs participate in the multistep acquisition of specific traits developed by tumor cells, which are referred to as "the hallmarks of cancer" defined by Hanahan and Weinberg. Moreover, as will be discussed, EVs play an important role in drug resistance, and these more recent advances may explain, at least in part, why pharmacological therapies are often ineffective. Finally, we discuss literature proposing the use of EVs for therapeutic and prognostic purposes in cancer.
Collapse
Affiliation(s)
- Renato Burgos-Ravanal
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - América Campos
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, UQ Centre for Clinical Research, Royal Brisbane and Women’s Hospital, The University of Queensland, Brisbane 4029, Australia
| | - Magda C. Díaz-Vesga
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Pontificia Universidad Javeriana de Cali, Cali 760008, Colombia
| | - María Fernanda González
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - Daniela León
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Lorena Lobos-González
- Centro de Medicina Regenerativa, Facultad de Medicina, Universidad del Desarrollo-Clínica Alemana, Santiago 7590943, Chile;
| | - Lisette Leyton
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| | - Marcelo J. Kogan
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Andrew F. G. Quest
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Programa de Biología Celular y Molecular, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (R.B.-R.); (A.C.); (M.C.D.-V.); (M.F.G.); (L.L.)
- Centro Avanzado para Estudios en Enfermedades Crónicas (ACCDIS), Santiago 8380453, Chile;
| |
Collapse
|
33
|
Palacios-Acedo AL, Mege D, Crescence L, Panicot-Dubois L, Dubois C. Cancer animal models in thrombosis research. Thromb Res 2021; 191 Suppl 1:S112-S116. [PMID: 32736767 DOI: 10.1016/s0049-3848(20)30407-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/07/2019] [Accepted: 12/14/2019] [Indexed: 12/11/2022]
Abstract
The cancer-thrombosis relationship has been established for decades, in both cancer biology and in the clinical signs and symptoms seen in cancer patients (thrombosis in cancer patients has been associated with a worse prognosis and survival). As the link between the pathologies becomes clearer, so does the need to develop models that enable researchers to study them simultaneously in vivo. Mouse models have often been used, and they have helped determine molecular pathways between cancer spread and thrombosis in humans. This review is a summary of the current literature that describes the use of cancer mouse models in thrombosis research. We included cancer models that are not yet used in thrombosis research, but that can positively impact this area of research in the near future. We describe the most commonly used techniques to generate thrombosis as well as the mouse strains and cancer cell types that are commonly used along with inoculation techniques. We endeavoured to create a compendium of the different mouse models that are beneficial for cancer-thrombosis research, as understanding these mechanisms is crucial for creating better and more effective treatments for thrombosis in cancer patients.
Collapse
Affiliation(s)
| | - Diane Mege
- Aix Marseille University, INSERM 1263, INRAE, C2VN, Marseille, France; Department of Digestive Surgery, Timone University Hospital, Marseille, France
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263, INRAE, C2VN, Marseille, France
| | | | - Christophe Dubois
- Aix Marseille University, INSERM 1263, INRAE, C2VN, Marseille, France.
| |
Collapse
|
34
|
Plasminogen activator inhibitor 1 and venous thrombosis in pancreatic cancer. Blood Adv 2021; 5:487-495. [PMID: 33496742 DOI: 10.1182/bloodadvances.2020003149] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/17/2020] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer patients have a high risk of venous thromboembolism (VTE). Plasminogen activator inhibitor 1 (PAI-1) inhibits plasminogen activators and increases the risk of thrombosis. PAI-1 is expressed by pancreatic tumors and human pancreatic cell lines. However, to date, there are no studies analyzing the association of active PAI-1 and VTE in pancreatic cancer patients. We investigated the association of active PAI-1 in plasma and VTE in pancreatic cancer patients. In addition, we determined if the presence of human pancreatic tumors expressing PAI-1 impairs venous thrombus resolution in mice. Plasma levels of active PAI-1 in patients with pancreatic cancer and mice bearing human tumors were determined by enzyme-linked immunosorbent assay. We measured PAI-1 expression in 5 different human pancreatic cancer cell lines and found that PANC-1 cells expressed the highest level. PANC-1 tumors were grown in nude mice. Venous thrombosis was induced by complete ligation of the inferior vena cava (IVC). Levels of active PAI-1 were independently associated with increased risk of VTE in patients with pancreatic cancer (subdistribution hazard ratio per doubling of levels: 1.39 [95% confidence interval, 1.09-1.78], P = .007). Mice bearing PANC-1 tumors had increased levels of both active human and active mouse PAI-1 and decreased levels of plasmin activity. Importantly, mice bearing PANC-1 tumors exhibited impaired venous thrombus resolution 8 days after IVC stasis compared with nontumor controls. Our results suggest that PAI-1 contributes to VTE in pancreatic cancer.
Collapse
|
35
|
Hisada Y, Moser B, Kawano T, Revenko AS, Crosby JR, Spronk HM, Mackman N. The Intrinsic Pathway does not Contribute to Activation of Coagulation in Mice Bearing Human Pancreatic Tumors Expressing Tissue Factor. Thromb Haemost 2021; 121:967-970. [PMID: 33498089 DOI: 10.1055/s-0040-1722193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Bernhard Moser
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Tomohiro Kawano
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Alexey S Revenko
- Ionis Pharmaceuticals, Inc., Antisense Drug Discovery, Carlsbad, California, United States
| | - Jeff R Crosby
- Ionis Pharmaceuticals, Inc., Antisense Drug Discovery, Carlsbad, California, United States
| | - Henri M Spronk
- Department of Internal Medicine and Biochemistry, Laboratory for Clinical Thrombosis and Hemostasis, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
36
|
Li Y, Yuan R, Ren T, Yang B, Miao H, Liu L, Li Y, Cai C, Yang Y, Hu Y, Jiang C, Xu Q, Zhang Y, Liu Y. Role of Sciellin in gallbladder cancer proliferation and formation of neutrophil extracellular traps. Cell Death Dis 2021; 12:30. [PMID: 33414368 PMCID: PMC7791032 DOI: 10.1038/s41419-020-03286-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
Apart from primary tumor development and metastasis, cancer-associated thrombosis is the second cause of cancer death in solid tumor malignancy. However, the mechanistic insight into the development of gallbladder cancer (GBC) and cancer-associated thrombosis remains unclear. This study aimed to investigate the mechanistic role of Sciellin (SCEL) in GBC cell proliferation and the development of venous thromboembolism. The expression level of SCEL was determined by immunohistochemical staining. Roles of SCEL in gallbladder cancer cell were determined by molecular and cell biology methods. SCEL was markedly upregulated in GBC and associated with advanced TNM stages and a poor prognosis. Furthermore, SCEL interacted with EGFR and stabilized EGFR expression that activates downstream PI3K and Akt pathway, leading to cell proliferation. In addition, SCEL induces tumor cell IL-8 production that stimulates the formation of neutrophil extracellular traps (NETs), accelerating thromboembolism. In xenografts, SCEL-expressing GBCs developed larger tumors and thrombosis compared with control cells. The present results indicate that SCEL promotes GBC cell proliferation and induces NET-associated thrombosis, thus serving as a potential therapeutic target.
Collapse
Affiliation(s)
- Yang Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ruiyan Yuan
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Tai Ren
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Bo Yang
- Department of General Surgery, First Affiliated Hospital of Wenzhou Medical University, Baixiang Road, Wenzhou, 325000, China
| | - Huijie Miao
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Liguo Liu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yongsheng Li
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Chen Cai
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yang Yang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yunping Hu
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Chengkai Jiang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qindie Xu
- School of Clinical Medicine, Shanghai University of Medicine & Health Sciences, 279 Zhouzhugong Road, Shanghai, 201318, China
| | - Yijian Zhang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Shanghai Research Center of Biliary Tract Disease, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
37
|
Grover SP, Olson TM, Cooley BC, Mackman N. Model-dependent contributions of FXII and FXI to venous thrombosis in mice. J Thromb Haemost 2020; 18:2899-2909. [PMID: 33094904 PMCID: PMC7693194 DOI: 10.1111/jth.15037] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/07/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The intrinsic pathway factors (F) XII and FXI have been shown to contribute to thrombosis in animal models. We assessed the role of FXII and FXI in venous thrombosis in three distinct mouse models. METHODS Venous thrombosis was assessed in mice genetically deficient for either FXII or FXI. Three models were used: the inferior vena cava (IVC) stasis, IVC stenosis, and femoral vein electrolytic injury models. RESULTS In the IVC stasis model, FXII and FXI deficiency did not affect the size of thrombi but their absence was associated with decreased levels of fibrin(ogen) and an increased level of the neutrophil extracellular trap marker citrullinated histone H3. In contrast, a deficiency of either FXII or FXI resulted in a significant and equivalent reduction in thrombus weight and incidence of thrombus formation in the IVC stenosis model. Thrombi formed in the IVC stenosis model contained significantly higher levels of citrullinated histone H3 compared with the thrombi formed in the IVC stasis model. Deletion of either FXII or FXI also resulted in a significant and equivalent reduction in both fibrin and platelet accumulation in the femoral vein electrolytic injury model. CONCLUSIONS Collectively, these data indicate that FXII and FXI contribute to the size of venous thrombosis in models with blood flow and thrombus composition in a stasis model. This study also demonstrates the importance of using multiple mouse models to assess the role of a given protein in venous thrombosis.
Collapse
Affiliation(s)
- Steven P. Grover
- Division of Hematology and OncologyDepartment of MedicineUNC Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Tatianna M. Olson
- Division of Hematology and OncologyDepartment of MedicineUNC Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Brian C. Cooley
- Department of Pathology and Laboratory MedicineMcAllister Heart InstituteUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Nigel Mackman
- Division of Hematology and OncologyDepartment of MedicineUNC Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| |
Collapse
|
38
|
Kim AS, Khorana AA, McCrae KR. Mechanisms and biomarkers of cancer-associated thrombosis. Transl Res 2020; 225:33-53. [PMID: 32645431 PMCID: PMC8020882 DOI: 10.1016/j.trsl.2020.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer-associated thrombosis is a leading cause of non-cancer death in cancer patients and is comprised of both arterial and venous thromboembolism (VTE). There are multiple risk factors for developing VTE, including cancer type, stage, treatment, and other medical comorbidities, which suggests that the etiology of thrombosis is multifactorial. While cancer-associated thrombosis can be treated with anticoagulation, benefits of therapy must be balanced with the increased bleeding risks seen in patients with cancer. Although risk models exist for primary and recurrent VTE, additional predictors are needed to improve model performance and discrimination of high-risk patients. This review will outline the diverse mechanisms driving thrombosis in cancer patients, as well as provide an overview of biomarkers studied in thrombosis risk and important considerations when selecting candidate biomarkers.
Collapse
Affiliation(s)
- Ann S Kim
- Taussig Cancer Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Alok A Khorana
- Taussig Cancer Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Keith R McCrae
- Taussig Cancer Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
39
|
Avisar A, Cohen M, Brenner B, Bronshtein T, Machluf M, Bar-Sela G, Aharon A. Extracellular Vesicles Reflect the Efficacy of Wheatgrass Juice Supplement in Colon Cancer Patients During Adjuvant Chemotherapy. Front Oncol 2020; 10:1659. [PMID: 32984039 PMCID: PMC7479215 DOI: 10.3389/fonc.2020.01659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Colorectal cancer (CC) is the third most common type of cancer, accounting for 10% of all cancer cases. Adjuvant chemotherapy is recommended in stages II–III CC. Wheatgrass juice (WGJ) from wheat seeds has high nutritional values, may induce synergistic benefits to chemotherapy and may attenuate chemotherapy-related side effects. Extracellular vesicles (EVs) are subcellular membrane blebs. EVs include exosomes (generated in the endosome, in size <150 nm) and microvesicles (shed from the plasma cell membrane) provide information on their parental cells and play a role in intercellular communication. We aimed to elucidate the effects of chemotherapy administration with supportive treatment of WGJ on CC patients’ EVs characteristics. Methods EVs were isolated from the blood samples of 15 healthy controls (HCs) and 50 CC patients post-surgery, treated by chemotherapy, with or without additional daily WGJ. Blood samples were taken before, during, and at the end of chemotherapy. EVs were characterized by size, concentration, membrane antigens and cytokine content using nanoparticle-tracking analysis, western blot, flow cytometry, and protein array methods. Results EVs were found to be similar by size and concentration with reduced levels of exosome markers (CD81) on samples at the end of combined treatment (chemotherapy and WGJ). Higher levels of endothelial EVs, which may indicate impairment of the vascular endothelial cells during treatment, were found in CC patients treated by chemotherapy only compared to those with chemotherapy and daily WGJ. Also, EVs thrombogenicity was lower in patients added WGJ compared to patients who had only chemotherapy (levels of tissue factor p = 0.029 and endothelial protein C receptor p = 0.005). Following treatments, levels of vascular endothelial growth factor receptors (VEGFR-1) and the majority of growth-factors/pro-inflammatory cytokines were higher in EVs of patients treated by chemotherapy only than in EVs obtained from patients with the combined treatment. Conclusion Daily consumption of WGJ during chemotherapy may reduce vascular damage and chemotherapy-related thrombogenicity, growth factors and cytokines, as reflected by the characteristics of patient’s EVs.
Collapse
Affiliation(s)
- Adva Avisar
- The Graduate Studies Authority, University of Haifa, Haifa, Israel
| | - Miri Cohen
- School of Social Work, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tomer Bronshtein
- The Lab for Cancer Drug Delivery & Cell Based Technologies, The Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Marcelle Machluf
- The Lab for Cancer Drug Delivery & Cell Based Technologies, The Faculty of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Gil Bar-Sela
- Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Cancer Center, Emek Medical Center, Afula, Israel
| | - Anat Aharon
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Hematology Research Laboratory, Hematology and Bone Marrow Transplantation, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Cancer cell-derived tissue factor-positive extracellular vesicles: biomarkers of thrombosis and survival. Curr Opin Hematol 2020; 26:349-356. [PMID: 31261175 DOI: 10.1097/moh.0000000000000521] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Tissue factor (TF) is released from cancer cells and tumors in the form of extracellular vesicles (EVs). This review summarizes our current knowledge of the mechanisms of release of TF-positive EVs (TF+EVs) from cancer cells and the effect of these TF+EVs on cultured endothelial cells. In addition, we will summarize the contribution of TF+EVs to thrombosis in mice, and the association between plasma EVTF activity and venous thrombosis as well as survival of cancer patients. RECENT FINDINGS The release of TF+EVs from cancer cells is regulated by multiple factors, including hypoxia, epithelial-mesenchymal transition, and various intracellular signaling pathways. Cancer cell-derived, TF+EVs confer procoagulant activity to endothelial cells and induce the expression of adhesion proteins and IL-8. In addition, they contribute to thrombosis by directly activating the coagulation system and by generating thrombin that activates platelets in mouse models. Finally, there is an association between EVTF activity and venous thrombosis in pancreatic cancer patients as well as mortality in cancer patients. SUMMARY Cancer cell-derived TF+EVs bind to and activate endothelial cells. In addition, they serve as biomarkers of survival of cancer patients and venous thrombosis in pancreatic cancer patients.
Collapse
|
41
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a Mouse Model of Venous Thrombosis. Arterioscler Thromb Vasc Biol 2020; 39:311-318. [PMID: 30786739 DOI: 10.1161/atvbaha.118.311818] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis. Leading experts in venous thrombosis research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of venous thrombosis. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of venous thrombosis. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
Affiliation(s)
- Jose A Diaz
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Prakash Saha
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brian Cooley
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Olivia R Palmer
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Steven P Grover
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Nigel Mackman
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill (B.C., S.P.G., N.M.)
| | - Thomas W Wakefield
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Peter K Henke
- From the Department of Surgery, Vascular Surgery, University of Michigan, Ann Arbor (J.A.D., O.R.P., T.W.W., P.K.H.)
| | - Alberto Smith
- Academic Department of Vascular Surgery, King's College London, UK (P.S., A.S.)
| | - Brajesh K Lal
- Department of Surgery, University of Maryland, College Park (B.K.L.)
| |
Collapse
|
42
|
Li D, Zhang X, Zhang H, Li X. Synergic effect of GPIBA and von Willebrand factor in pathogenesis of deep vein thrombosis. Vascular 2020; 28:309-313. [PMID: 31902309 DOI: 10.1177/1708538119896446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES In cardiovascular disease, deep vein thrombosis is one of the vital symptoms causing pulmonary thromboembolism. However, the pathogenesis of deep vein thrombosis is still not clear. One of the critical factors leading to deep vein thrombosis is the platelet aggregation that is mediated by a set of key genes including platelet membrane protein coded by platelet glycoprotein Ib alpha chain (GPIBA). METHODS Deep vein thrombosis model was established according to the previous protocol, and venous blood and thrombi were collected for further analysis. RESULTS The dynamic changes of GPIBA and coagulation factor, von Willebrand factor, were observed in deep vein thrombosis models. Meanwhile, critical proteins participating in adhesion and binding of platelets such as epithelial membrane protein 2 (EMP2), vascular cell adhesion protein 1 (VCAM1), immunoreceptor tyrosine-based activation motif 1 (ITAM1), integrin subunit alpha M (ITGAM), or fibronectin were also differentially expressed in deep vein thrombosis models. CONCLUSIONS Application of heparin could reverse these dynamic changes in deep vein thrombosis models. Thus, we explained the potential synergic role of GPIBA and von Willebrand factor in regulating the occurrence of deep vein thrombosis and provide therapeutic target against cardiovascular disease.
Collapse
Affiliation(s)
- Da Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Soochow University, Suzhou, China.,Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaosong Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Honggang Zhang
- Department of Vascular Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital, Soochow University, Suzhou, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
43
|
Hisada Y, Grover SP, Maqsood A, Houston R, Ay C, Noubouossie DF, Cooley BC, Wallén H, Key NS, Thålin C, Farkas ÁZ, Farkas VJ, Tenekedjiev K, Kolev K, Mackman N. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020; 105:218-225. [PMID: 31048354 PMCID: PMC6939515 DOI: 10.3324/haematol.2019.217083] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/24/2019] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is associated with a high incidence of venous thromboembolism. Neutrophils have been shown to contribute to thrombosis in part by releasing neutrophil extracellular traps (NET). A recent study showed that increased plasma levels of the NET biomarker, citrullinated histone H3 (H3Cit), are associated with venous thromboembolism in patients with pancreatic and lung cancer but not in those with other types of cancer, including breast cancer. In this study, we examined the contribution of neutrophils and NET to venous thrombosis in nude mice bearing human pancreatic tumors. We found that tumor-bearing mice had increased circulating neutrophil counts and levels of granulocyte-colony stimulating factor, neutrophil elastase, H3Cit and cell-free DNA compared with controls. In addition, thrombi from tumor-bearing mice contained increased levels of the neutrophil marker Ly6G, as well as higher levels of H3Cit and cell-free DNA. Thrombi from tumor-bearing mice also had denser fibrin with thinner fibers consistent with increased thrombin generation. Importantly, either neutrophil depletion or administration of DNase I reduced the thrombus size in tumor-bearing but not in control mice. Our results, together with clinical data, suggest that neutrophils and NET contribute to venous thrombosis in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yohei Hisada
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven P Grover
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anaum Maqsood
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Reaves Houston
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cihan Ay
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Denis F Noubouossie
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brian C Cooley
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Håkan Wallén
- Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nigel S Key
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charlotte Thålin
- Department of Clinical Sciences, Danderyd Hospital, Division of Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ádám Z Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika J Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Kiril Tenekedjiev
- Australian Maritime College, University of Tasmania, Launceston, Australia
- Department of Information Technology, Nikola Vaptsarov Naval Academy, Varna, Bulgaria
| | - Krasimir Kolev
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
44
|
Hisada Y, Mackman N. Update from the laboratory: mechanistic studies of pathways of cancer-associated venous thrombosis using mouse models. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:182-186. [PMID: 31808871 PMCID: PMC6913477 DOI: 10.1182/hematology.2019000025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Cancer patients have an increased risk of venous thromboembolism (VTE). The rate of VTE varies with cancer type, with pancreatic cancer having one of the highest rates, suggesting that there are cancer type-specific mechanisms of VTE. Risk assessment scores, such as the Khorana score, have been developed to identify ambulatory cancer patients at high risk of VTE. However, the Khorana score performed poorly in discriminating pancreatic cancer patients at risk of VTE. Currently, thromboprophylaxis is not recommended for cancer outpatients. Recent clinical trials showed that factor Xa (FXa) inhibitors reduced VTE in high-risk cancer patients but also increased major bleeding. Understanding the mechanisms of cancer-associated thrombosis should lead to the development of safer antithrombotic drugs. Mouse models can be used to study the role of different prothrombotic pathways in cancer-associated thrombosis. Human and mouse studies support the notion that 2 prothrombotic pathways contribute to VTE in pancreatic cancer patients: tumor-derived, tissue factor-positive (TF+) extracellular vesicles (EVs), and neutrophils and neutrophil extracellular traps (NETs). In pancreatic cancer patients, elevated levels of plasma EVTF activity and citrullinated histone H3 (H3Cit), a NET biomarker, are independently associated with VTE. We observed increased levels of circulating tumor-derived TF+ EVs, neutrophils, cell-free DNA, and H3Cit in nude mice bearing human pancreatic tumors. Importantly, inhibition of tumor-derived human TF, depletion of neutrophils, or administration of DNAse I to degrade cell-free DNA (including NETs) reduced venous thrombosis in tumor-bearing mice. These studies demonstrate that tumor-derived TF+ EVs, neutrophils, and cell-free DNA contribute to venous thrombosis in a mouse model of pancreatic cancer.
Collapse
Affiliation(s)
- Yohei Hisada
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
45
|
Isolated tumour microparticles induce endothelial microparticle release in vitro. Blood Coagul Fibrinolysis 2019; 31:35-42. [PMID: 31789658 DOI: 10.1097/mbc.0000000000000876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
: Cancer induces a hypercoagulable state, resulting in an increased risk of venous thromboembolism. One of the mechanisms driving this is tissue factor (TF) production by the tumour, released in small lipid bound microparticles. We have previously demonstrated that tumour cell line media-induced procoagulant changes in HUVEC. The aim of this study was to investigate the effect of tumour microparticles and recombinant human TF (rhTF) on the endothelium. Procoagulant microparticles from the PANC-1 cell line were harvested by ultrafiltration. HUVEC were then incubated with these procoagulant microparticles or rhTF. Flow cytometry was used to investigate the effect of endothelial cell surface protein expression and microparticle release. Microparticles but not soluble TF was responsible for the procoagulant activity of cell-free tumour media. We also demonstrated an increase in endothelial microparticle release with exposure to tumour microparticles, with a positive linear relationship observed (R = 0.6630 P ≤ 0.0001). rhTF did not induce any of the changes observed with microparticles. Here we demonstrate that procoagulant activity of tumour cell line media is dependent on microparticles, and that exposure of endothelial cells to these microparticles results in an increase in microparticle release from HUVEC. This suggests a mechanism of transfer of procoagulant potential from the cancer to the remote endothelium.
Collapse
|
46
|
The relationship between pancreatic cancer and hypercoagulability: a comprehensive review on epidemiological and biological issues. Br J Cancer 2019; 121:359-371. [PMID: 31327867 PMCID: PMC6738049 DOI: 10.1038/s41416-019-0510-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
It has long been recognised that pancreatic cancer induces a hypercoagulable state that may lead to clinically apparent thrombosis. Although the relationship between pancreatic cancer and hypercoagulability is well described, the underlying pathological mechanism(s) and the interplay between these pathways remain a matter of intensive study. This review summarises existing data on epidemiology and pathogenesis of thrombotic complications in pancreatic cancer with a particular emphasis on novel pathophysiological pathways. Pancreatic cancer is characterised by high tumoural expression of tissue factor, activation of leukocytes with the release of neutrophil extracellular traps, the dissemination of tumour-derived microvesicles that promote hypercoagulability and increased platelet activation. Furthermore, other coagulation pathways probably contribute to these processes, such as those that involve heparanase, podoplanin and hypofibrinolysis. In the era in which heparin and its derivatives—the currently recommended therapy for cancer-associated thrombosis—might be superseded by direct oral anticoagulants, novel data from mouse models of cancer-associated thrombosis suggest the possibility of future personalised therapeutic approaches. In this dynamic era for cancer-associated thrombosis, the discovery of novel prothrombotic and proinflammatory mechanisms will potentially uncover pharmacological targets to prevent and treat thrombosis without adversely affecting haemostasis.
Collapse
|
47
|
Novel Aspects of Extracellular Vesicles as Mediators of Cancer-Associated Thrombosis. Cells 2019; 8:cells8070716. [PMID: 31337034 PMCID: PMC6679024 DOI: 10.3390/cells8070716] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
The establishment of prothrombotic states during cancer progression is well reported but the precise mechanisms underlying this process remain elusive. A number of studies have implicated the presence of the clotting initiator protein, tissue factor (TF), in circulating tumor-derived extracellular vesicles (EVs) with thrombotic manifestations in certain cancer types. Tumor cells, as well as tumor-derived EVs, may activate and promote platelet aggregation by TF-dependent and independent pathways. Cancer cells and their secreted EVs may also facilitate the formation of neutrophil extracellular traps (NETs), which may contribute to thrombus development. Alternatively, the presence of polyphosphate (polyP) in tumor-derived EVs may promote thrombosis through a TF-independent route. We conclude that the contribution of EVs to cancer coagulopathy is quite complex, in which one or more mechanisms may take place in a certain cancer type. In this context, strategies that could attenuate the crosstalk between the proposed pro-hemostatic routes could potentially reduce cancer-associated thrombosis.
Collapse
|
48
|
Diaz JA, Saha P, Cooley B, Palmer OR, Grover SP, Mackman N, Wakefield TW, Henke PK, Smith A, Lal BK. Choosing a mouse model of venous thrombosis: a consensus assessment of utility and application. J Thromb Haemost 2019; 17:699-707. [PMID: 30927321 DOI: 10.1111/jth.14413] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Murine models are widely used valuable tools to study deep vein thrombosis (VT). Leading experts in VT research came together through the American Venous Forum to develop a consensus on maximizing the utility and application of available mouse models of VT. In this work, we provide an algorithm for model selection, with discussion of the advantages, disadvantages, and applications of the main mouse models of VT. Additionally, we provide a detailed surgical description of the models with guidelines to validate surgical technique.
Collapse
|
49
|
Thrombin Generation and Cancer: Contributors and Consequences. Cancers (Basel) 2019; 11:cancers11010100. [PMID: 30654498 PMCID: PMC6356447 DOI: 10.3390/cancers11010100] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
The high occurrence of cancer-associated thrombosis is associated with elevated thrombin generation. Tumour cells increase the potential for thrombin generation both directly, through the expression and release of procoagulant factors, and indirectly, through signals that activate other cell types (including platelets, leukocytes and erythrocytes). Furthermore, cancer treatments can worsen these effects. Coagulation factors, including tissue factor, and inhibitors of coagulation are altered and extracellular vesicles (EVs), which can promote and support thrombin generation, are released by tumour and other cells. Some phosphatidylserine-expressing platelet subsets and platelet-derived EVs provide the surface required for the assembly of coagulation factors essential for thrombin generation in vivo. This review will explore the causes of increased thrombin production in cancer, and the availability and utility of tests and biomarkers. Increased thrombin production not only increases blood coagulation, but also promotes tumour growth and metastasis and as a consequence, thrombin and its contributors present opportunities for treatment of cancer-associated thrombosis and cancer itself.
Collapse
|
50
|
Abstract
Venous thromboembolism is known to be associated with an increase in morbidity and mortality in patients with malignancy. Predictive laboratory biomarkers of venous thromboembolism (VTE) have long been sought after to improve outcomes and help guide clinical decision making. Previously studied biomarkers include C reactive protein (CRP), tissue factor expressing microparticles (TF MP), D-dimer, soluble P-selectin (sP-selectin), plasminogen activator inhibitor 1 (PAI-1), factor VIII, platelet count, and leukocyte counts. This chapter will focus on these possible biomarkers for cancer-associated thrombosis (CAT) with particular emphasis on the pathophysiology behind thrombosis formation as well as data from clinical studies in patients with malignancy. The incorporation of the above biomarkers into risk assessment tools to predict CAT will also be reviewed, as will risk factors for recurrent VTE in patients with malignancy. Further studies are ongoing to develop readily available biomarkers that can be incorporated into future risk assessment models with the goal of reducing morbidity and mortality due to cancer-associated thrombosis.
Collapse
Affiliation(s)
- Anjlee Mahajan
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA.
| | - Ted Wun
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA
- UC Davis School of Medicine, Clinical and Translational Sciences Center (CTSC), Sacramento, USA
| |
Collapse
|