1
|
Xu W, Tan X, Li ML, Xu H, Villegas J, Fu H. Von Willebrand factor and hematogenous cancer metastasis under flow. Front Cell Dev Biol 2024; 12:1435718. [PMID: 39282473 PMCID: PMC11401050 DOI: 10.3389/fcell.2024.1435718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
Hematogenous metastasis involves cancer cell migration to different locations from the primary tumor through the blood circulation. Von Willebrand factor (VWF) has been shown to play an important role in tumor cell adhesion to and extravasation from the endothelial cell lining of blood vessel walls during cancer metastasis. VWF may contribute to this process by interacting with tumor cells, endothelial cells, and platelets through various cell membrane receptors, such as platelet glycoprotein (GP)Ibα, P-selectin, ανβ3 and αIIbβ3 integrins, and glycocalyx. Blood flow can mechanically extend and activate VWF to bind platelets and associate intermolecularly with other VWF molecules in plasma or on the surface of endothelial cells, cancer cells, or platelets. This suggests a mechanoregulatory role of VWF in mediating the interactions between VWF and these cells to promote cancer cell adhesion to blood vessels. In this review, we will summarize the current knowledge of VWF function and the role of hydrodynamic forces in hematogenous cancer metastasis.
Collapse
Affiliation(s)
- Wenxuan Xu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Xi Tan
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Morgan L Li
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Hanzhi Xu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Jasmine Villegas
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Hongxia Fu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
- Department of Bioengineering, University of Washington, Seattle, WA, United States
- Bloodworks Research Institute, Seattle, WA, United States
| |
Collapse
|
2
|
Oblitas CM, Demelo-Rodríguez P, López-Rubio M, Lago-Rodríguez MO, García-Gámiz M, Zamora-Trillo A, Alvarez-Sala Walther LA, García-Martínez R, Galeano-Valle F. Evaluation of soluble P-selectin as a predictive biomarker in acute symptomatic pulmonary embolism: Insights from a prospective observational study. Eur J Haematol 2024; 113:201-207. [PMID: 38654526 DOI: 10.1111/ejh.14218] [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: 01/15/2024] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Soluble P-selectin (sP-selectin) has been proposed as a potential biomarker for venous thromboembolism (VTE) diagnosis with interesting results. However, its role in predicting early mortality in pulmonary embolism (PE) remains unexplored. METHODS This observational, prospective, single-center study enrolled consecutive patients aged 18 or older with confirmed acute symptomatic PE and no prior anticoagulation. The study aims to assess the prognostic capacity of sP-selectin measured at the time of PE diagnosis for short-term mortality and major bleeding. RESULTS A total of 196 patients, with a mean age of 69.1 years (SD 17), were included, of whom 52.6% were male. Within 30 days, 9.7% of patients (n = 19) died, and 5.1% (n = 10) suffered major bleeding. PE risk stratification revealed 4.6% (n = 9) with high-risk PE, 34.7% (n = 68) with intermediate-high-risk PE, 38.3% (n = 75) with intermediate-low-risk PE, and 22.5% (n = 44) with low-risk PE according to the European Society of Cardiology score. Mean plasma sP-selectin levels were comparable between survivors and non-survivors (489.7 ng/mL ±63 vs. 497.3 ng/mL ±51; p = .9). The ROC curve for 30-day all-cause mortality and major bleeding yielded an AUC of 0.49 (95% CI 0.36-0.63) and 0.46 (95% CI 0.24-0.68), respectively. Multivariate and survival analyses were precluded due to lack of significance. CONCLUSIONS sP-selectin was not useful for predicting short-term mortality or major bleeding in patients with acute symptomatic pulmonary embolism. Further studies are required to clarify the role of sP-selectin in VTE, particularly in prognosticating PE outcomes.
Collapse
Affiliation(s)
- Crhistian-Mario Oblitas
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Pablo Demelo-Rodríguez
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Marina López-Rubio
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Marta-Olimpia Lago-Rodríguez
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Mercedes García-Gámiz
- Department of Clinical Biochemistry, General University Hospital Gregorio Marañón, Madrid, Spain
| | - Angielys Zamora-Trillo
- Department of Clinical Biochemistry, General University Hospital Gregorio Marañón, Madrid, Spain
| | - Luis-Antonio Alvarez-Sala Walther
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| | - Rita García-Martínez
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
| | - Francisco Galeano-Valle
- Venous Thromboembolism Unit. Internal Medicine Department, General University Hospital Gregorio Marañón, Madrid, Spain
- School of Medicine, University Complutense of Madrid, Madrid, Spain
- Lipids and Cardiovascular Risk Unit, Sanitary Research Institute Gregorio Marañón, Madrid, Spain
| |
Collapse
|
3
|
Kast RE. IC Regimen: Delaying Resistance to Lorlatinib in ALK Driven Cancers by Adding Repurposed Itraconazole and Cilostazol. Cells 2024; 13:1175. [PMID: 39056757 PMCID: PMC11274432 DOI: 10.3390/cells13141175] [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: 06/21/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Lorlatinib is a pharmaceutical ALK kinase inhibitor used to treat ALK driven non-small cell lung cancers. This paper analyses the intersection of past published data on the physiological consequences of two unrelated drugs from general medical practice-itraconazole and cilostazol-with the pathophysiology of ALK positive non-small cell lung cancer. A conclusion from that data analysis is that adding itraconazole and cilostazol may make lorlatinib more effective. Itraconazole, although marketed worldwide as a generic antifungal drug, also inhibits Hedgehog signaling, Wnt signaling, hepatic CYP3A4, and the p-gp efflux pump. Cilostazol, marketed worldwide as a generic thrombosis preventative drug, acts by inhibiting phosphodiesterase 3, and, by so doing, lowers platelets' adhesion, thereby partially depriving malignant cells of the many tumor trophic growth factors supplied by platelets. Itraconazole may enhance lorlatinib effectiveness by (i) reducing or stopping a Hedgehog-ALK amplifying feedback loop, by (ii) increasing lorlatinib's brain levels by p-gp inhibition, and by (iii) inhibiting growth drive from Wnt signaling. Cilostazol, surprisingly, carries minimal bleeding risk, lower than that of aspirin. Risk/benefit assessment of the combination of metastatic ALK positive lung cancer being a low-survival disease with the predicted safety of itraconazole-cilostazol augmentation of lorlatinib favors a trial of this drug trio in ALK positive lung cancer.
Collapse
|
4
|
Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, Assis J, Pereira D, Medeiros R. Haemostatic Gene Expression in Cancer-Related Immunothrombosis: Contribution for Venous Thromboembolism and Ovarian Tumour Behaviour. Cancers (Basel) 2024; 16:2356. [PMID: 39001418 PMCID: PMC11240748 DOI: 10.3390/cancers16132356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Ovarian cancer (OC) is the deadliest gynaecological malignancy. Identifying new prognostic biomarkers is an important research field. Haemostatic components together with leukocytes can drive cancer progression while increasing the susceptibility to venous thromboembolism (VTE) through immunothrombosis. Unravelling the underlying complex interactions offers the prospect of uncovering relevant OC prognostic biomarkers, predictors of cancer-associated thrombosis (CAT), and even potential targets for cancer therapy. Thus, this study evaluated the expression of F3, F5, F8, F13A1, TFPI1, and THBD in peripheral blood cells (PBCs) of 52 OC patients. Those with VTE after tumour diagnosis had a worse overall survival (OS) compared to their counterparts (mean OS of 13.8 ± 4.1 months and 47.9 ± 5.7 months, respectively; log-rank test, p = 0.001). Low pre-chemotherapy F3 and F8 expression levels were associated with a higher susceptibility for OC-related VTE after tumour diagnosis (χ2, p < 0.05). Regardless of thrombogenesis, patients with low baseline F8 expression had a shorter progression-free survival (PFS) than their counterparts (adjusted hazard ratio (aHR) = 2.54; p = 0.021). Among those who were not under platelet anti-aggregation therapy, low F8 levels were also associated with a shorter OS (aHR = 6.16; p = 0.006). Moving forward, efforts should focus on external validation in larger cohorts.
Collapse
Affiliation(s)
- Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Joana Assis
- Clinical Research Unit, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto. CCC), 4200-072 Porto, Portugal;
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/Pathology and Laboratory Medicine Dep., Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), 4200-072 Porto, Portugal;
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
5
|
Tatsumi K. The pathogenesis of cancer-associated thrombosis. Int J Hematol 2024; 119:495-504. [PMID: 38421488 DOI: 10.1007/s12185-024-03735-x] [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: 12/04/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Patients with cancer have a higher risk of venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), compared to the general population. Cancer-associated thrombosis (CAT) is a thrombotic event that occurs as a complication of cancer or cancer therapy. Major factors determining VTE risk in cancer patients include not only treatment history and patient characteristics, but also cancer type and site. Cancer types can be broadly divided into three groups based on VTE risk: high risk (pancreatic, ovarian, brain, stomach, gynecologic, and hematologic), intermediate risk (colon and lung), and low risk (breast and prostate). This implies that the mechanism of VTE differs between cancer types and that specific VTE pathways may exist for different cancer types. This review summarizes the specific pathways that contribute to VTE in cancer patients, with a particular focus on leukocytosis, neutrophil extracellular traps (NETs), tissue factor (TF), thrombocytosis, podoplanin (PDPN), plasminogen activator inhibitor-1 (PAI-1), the intrinsic coagulation pathway, and von Willebrand factor (VWF).
Collapse
Affiliation(s)
- Kohei Tatsumi
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8521, Japan.
| |
Collapse
|
6
|
Yan AR, Samarawickrema I, Naunton M, Peterson GM, Yip D, Newman P, Mortazavi R. Models for predicting venous thromboembolism in ambulatory patients with lung cancer: A systematic review and meta-analysis. Thromb Res 2024; 234:120-133. [PMID: 38215613 DOI: 10.1016/j.thromres.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/29/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024]
Abstract
AIMS The incidence of venous thromboembolism (VTE) in patients with lung cancer is relatively high, and risk stratification models are vital for the targeted application of thromboprophylaxis. We aimed to review VTE risk prediction models that have been developed in patients with lung cancer and evaluated their performance. METHODS AND RESULTS Twenty-four eligible studies involving 123,493 patients were included. The pooled incidence of VTE within 12 months was 11 % (95 % CI 8 %-14 %). With the identified four VTE risk assessment tools, meta-analyses did not show a significant discriminatory capability of stratifying VTE risk for Khorana, PROTECHT and CONKO scores. The pooled sensitivity and specificity of the Khorana score were 24 % (95 % CI 11 %-44 %) and 84 % (95 % CI 73 %-91 %) at the 3-point cut-off, and 43 % (95 % CI 35 %-52 %) and 61 % (95 % CI 52 %-69 %) at the 2-point cut-off. However, a COMPASS-CAT score of ≥ 7 points indicated a significantly high VTE risk, with a RR of 4.68 (95 % CI 1.05-20.80). CONCLUSIONS The Khorana score lacked discriminatory capability in identifying patients with lung cancer at high VTE risk, regardless of the cut-off value. The COMPASS-CAT score had better performance, but further validation is needed. The results indicate the need for robust VTE risk assessment tools specifically designed and validated for lung cancer patients. Future research should include relevant biomarkers as important predictors and consider the combined use of risk tools. PROSPERO registration number: CRD42021245907.
Collapse
Affiliation(s)
- Ann-Rong Yan
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra, ACT, Australia.
| | | | - Mark Naunton
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra, ACT, Australia.
| | - Gregory M Peterson
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra, ACT, Australia; College of Health and Medicine, University of Tasmania, TAS, Australia.
| | - Desmond Yip
- ANU School of Medicine and Psychology, Australian National University, Canberra, ACT, Australia; Department of Medical Oncology, The Canberra Hospital, Garran, ACT, Australia.
| | - Phillip Newman
- Research Institute for Sport and Exercise, Faculty of Health, University of Canberra, Canberra, ACT, Australia.
| | - Reza Mortazavi
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra, ACT, Australia.
| |
Collapse
|
7
|
Drăgan A, Drăgan AŞ. Novel Insights in Venous Thromboembolism Risk Assessment Methods in Ambulatory Cancer Patients: From the Guidelines to Clinical Practice. Cancers (Basel) 2024; 16:458. [PMID: 38275899 PMCID: PMC10813930 DOI: 10.3390/cancers16020458] [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: 12/07/2023] [Revised: 01/07/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Many cancer patients will experience venous thromboembolism (VTE) at some stage, with the highest rate in the initial period following diagnosis. Novel cancer therapies may further enhance the risk. VTE in a cancer setting is associated with poor prognostic, a decreased quality of life, and high healthcare costs. If thromboprophylaxis in hospitalized cancer patients and perioperative settings is widely accepted in clinical practice and supported by the guidelines, it is not the same situation in ambulatory cancer patient settings. The guidelines do not recommend primary thromboprophylaxis, except in high-risk cases. However, nowadays, risk stratification is still challenging, although many tools have been developed. The Khrorana score remains the most used method, but it has many limits. This narrative review aims to present the current relevant knowledge of VTE risk assessment in ambulatory cancer patients, starting from the guideline recommendations and continuing with the specific risk assessment methods and machine learning models approaches. Biomarkers, genetic, and clinical features were tested alone or in groups. Old and new models used in VTE risk assessment are exposed, underlining their clinical utility. Imaging and biomolecular approaches to VTE screening of outpatients with cancer are also presented, which could help clinical decisions.
Collapse
Affiliation(s)
- Anca Drăgan
- Department of Cardiovascular Anaesthesiology and Intensive Care, Emergency Institute for Cardiovascular Diseases “Prof. Dr. C C Iliescu”, 258 Fundeni Road, 022328 Bucharest, Romania
| | - Adrian Ştefan Drăgan
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| |
Collapse
|
8
|
He X, Wei SN, Qin WW, Geng N, Li B, Song S, Wang P. Evaluating the effect of immune checkpoint inhibitors on venous thromboembolism in non-small cell lung cancer patients. Expert Rev Hematol 2023; 16:1135-1142. [PMID: 37883026 DOI: 10.1080/17474086.2023.2276209] [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: 04/09/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023]
Abstract
OBJECTIVE Currently, immune checkpoint inhibitors (ICIs) therapy is one of the main methods of treatment in non-small cell lung cancer (NSCLC). This study aimed to explore the risk factors of VTE and evaluate the effect of ICIs on VTE in patients with NSCLC. RESEARCH DESIGN AND METHODS We retrospectively studied patients with NSCLC who were divided into VTE group and without VTE (Non-VTE) group. We identified the risk factors of VTE in NSCLC patients and evaluated the effect of ICIs on VTE in NSCLC patients. RESULTS We found that clinical stage III-IV (P = 0.015) and Khorana score (KS) ≥ 2 (P = 0.047) were independent risk factors for the occurrence of VTE in NSCLC, and treatment with ICIs reduced the risk of VTE occurrence (P = 0.028). There were no differences of survival rates in the 12-month (P = 0.449), 24-month (P = 0.412), or 36-month (P = 0.315) between the VTE and non-VTE groups. History of anti-angiogenic therapy (P = 0.033) and chronic obstructive pulmonary disease (COPD) (P = 0.046) were independent risk factors for VTE in NSCLC patients who were treated with ICIs. CONCLUSION This study suggests that we should strengthen anticoagulant therapy when using ICIs for NSCLC patients with a history of anti-angiogenic therapy and COPD.
Collapse
Affiliation(s)
- Xin He
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Shan-Na Wei
- Department of Pulmonary and Critical Care Medicine, Hebei PetroChina Center Hospital, Langfang, P.R. China
| | - Wen-Wen Qin
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Nan Geng
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Bin Li
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Shan Song
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Ping Wang
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
9
|
Xiong W, Guo X, Du H, Xu M, Zhao Y. Management of venous thromboembolism in patients with lung cancer: a state-of-the-art review. BMJ Open Respir Res 2023; 10:10/1/e001493. [PMID: 37068846 PMCID: PMC10111887 DOI: 10.1136/bmjresp-2022-001493] [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: 10/06/2022] [Accepted: 03/31/2023] [Indexed: 04/19/2023] Open
Abstract
Venous thromboembolism (VTE) is common and life-threatening in patients with lung cancer. Management of VTE is critical for patients with lung cancer. Risk assessment, thromboprophylaxis and treatment of VTE constitute the core issues of VTE management in patients with lung cancer. Although its overall principles should follow recommendations in authoritative guidelines, VTE management in patients with lung cancer may be slightly special in some specific aspects. Despite the extensive validation of Khorana score for patients with all cancer types, its value in VTE risk assessment of patients with lung cancer is controversial. It is important to determine the VTE risk assessment score that can accurately and specifically assess the VTE risk of patients with lung cancer. Clinical practice patterns of thromboprophylaxis may vary by cancer types, since different sites of cancer may have different levels of VTE risk. To understand the thromboprophylaxis specific for lung cancer is of vital importance for patients with lung cancer. Although it is essential to comply with authoritative guidelines, the duration and timing of initiation of thromboprophylaxis in surgical patients with lung cancer may need further study. Taken together, the purpose of this review is to provide an overview of state-of-the-art VTE stewardship specific for patients with lung cancer.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Pulmonary and Critical Care Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuejun Guo
- Department of Pulmonary and Critical Care Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - He Du
- Department of Medical Oncology, Tongji University Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Mei Xu
- North Bund Community Health Service Center, Hongkou District, Shanghai, China
| | - Yunfeng Zhao
- Department of Pulmonary and Critical Care Medicine, Shanghai Punan Hospital, Shanghai, China
| |
Collapse
|
10
|
Sanfilippo KM, Wang TF, Carrier M, Falanga A, Gage BF, Khorana AA, Maraveyas A, Soff GA, Wells PS, Zwicker JI. Standardization of risk prediction model reporting in cancer-associated thrombosis: Communication from the ISTH SSC subcommittee on hemostasis and malignancy. J Thromb Haemost 2022; 20:1920-1927. [PMID: 35635332 DOI: 10.1111/jth.15759] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/27/2022]
Abstract
Since the development of the Khorana score to predict risk of cancer-associated venous thromboembolism (VTE), many modified and de novo risk prediction models (RPMs) have been proposed. Comparison of the prognostic performance across models requires comprehensive reporting and standardized methods for model development, validation and evaluation. To improve the standardization of RPM reporting, the Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD) tool was published in 2015. To better understand the quality of reporting and development of RPMs for cancer-associated VTE, we performed a literature search of published RPMs and assessed each model using the TRIPOD checklist. Our results yielded 29 RPMs for which 30 items were evaluated. There was a non-significant (p = 0.15) improvement in reporting of the 30 items in the post-TRIPOD era (81%) versus the pre-TRIPOD era (75%). Of seven items (title, sample size, missing data handling, baseline demographics, methods and results for model performance, and supplemental resources) with the lowest reporting in the pre-TRIPOD era (<70%), there was an average improvement of 22% in the post-TRIPOD era. Only two of the 22 studies published in the post-TRIPOD era acknowledged compliance with TRIPOD. Informed by the results of this assessment, the Scientific and Standardization Committee (SSC) Subcommittee on Hemostasis & Malignancy of the International Society on Thrombosis and Hemostasis (ISTH) advocates for standardization of four key elements of RPMs for cancer-associated VTE: (1) inclusion of the TRIPOD checklist, (2) clear definition of the derivation population, with justification of sample size, (3) clear definition of predictors, and (4) external validation prior to implementation.
Collapse
Affiliation(s)
- Kristen M Sanfilippo
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- John Cochran Saint Louis Veterans Administration Medical Center, Saint Louis, Missouri, USA
| | - Tzu-Fei Wang
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Marc Carrier
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Anna Falanga
- Department of Medicine and Surgery, University of Milan Bicocca, Milan, Italy
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Brian F Gage
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Alok A Khorana
- Taussig Cancer Institute, Cleveland Clinic and Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Anthony Maraveyas
- Faculty of Health Sciences, Joint Centre for Cancer Studies, The Hull York Medical School, Castle Hill Hospital, Hull, UK
| | - Gerald A Soff
- Department of Medicine, University of Miami Health System/Sylvester Comprehensive Cancer Center, Miami, Florida, USA
| | - Phillip S Wells
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Jeffrey I Zwicker
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Purdy M, Obi A, Myers D, Wakefield T. P- and E- selectin in venous thrombosis and non-venous pathologies. J Thromb Haemost 2022; 20:1056-1066. [PMID: 35243742 PMCID: PMC9314977 DOI: 10.1111/jth.15689] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022]
Abstract
Venous thromboembolism is a very common and costly health problem worldwide. Anticoagulant treatment for VTE is imperfect: all have the potential for significant bleeding, and none prevent the development of post thrombotic syndrome after deep vein thrombosis or chronic thromboembolic pulmonary hypertension after pulmonary embolism. For these reasons, alternate forms of therapy with improved efficacy and decreased bleeding are needed. Selectins are a family (P-selectin, E-selectin, L-selectin) of glycoproteins that facilitate and augment thrombosis, modulating neutrophil, monocyte, and platelet activity. P- and E-selectin have been investigated as potential biomarkers for thrombosis. Inhibition of P-selectin and E-selectin decrease thrombosis and vein wall fibrosis, with no increase in bleeding. Selectin inhibition is a promising avenue of future study as either a stand-alone treatment for VTE or as an adjunct to standard anticoagulation therapies.
Collapse
Affiliation(s)
- Megan Purdy
- University of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Andrea Obi
- Section of Vascular SurgeryDepartment of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Daniel Myers
- Section of Vascular SurgeryDepartment of SurgeryUniversity of MichiganAnn ArborMichiganUSA
- Unit for Laboratory Animal Medicine and Section of Vascular SurgeryDepartment of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Thomas Wakefield
- Section of Vascular SurgeryDepartment of SurgeryUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
12
|
Wu Y, Wan S, Chen Y, Fan J, Li Y, Wang T, Yuan Z, Yang Q, Qin H, Xu J, Zhang J. Biomimetic lipidic nanovectors for effective asparaginase supramolecule delivery. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102518. [PMID: 35032628 DOI: 10.1016/j.nano.2022.102518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 12/02/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022]
Abstract
Effectiveness of enzyme therapy is limited by enzyme drawbacks such as short half-life, low bioavailability and high immunogenicity. We loaded asparaginase (Aase) into hydroxypropyl- or sulfonbutylether-beta cyclodextrin to form supramolecular amphiphilic molecules by self-assembly followed by entrapment inside the cores of two biomimetic lipidic nanovectors (AS-XLNs). Supramolecular structure was simulated by molecular docking. AS-XLNs maintained superior activity through isolating Aase from external environment due to docking with cyclodextrin and coating with biomimetic membrane. Fluorescent probes and computational simulations were used to reveal possible interactions between serum albumin/trypsin and Aase/nanovector membrane components which were partly responsible for enhanced bioavailability and bioactivity of AS-XLNs compared to Aase. AS-XLNs significantly increased cytotoxicity against pulmonary tumor cells due to synergistic effects of Aase and nanovector membrane components (killing tumor cells through apoptosis induced by asparagine depletion and autophagy inhibition or via targets such as vascular endothelial growth factor A, alpha-amylase, p-selectin or androgen receptor).
Collapse
Affiliation(s)
- Yan Wu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Shengli Wan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Yun Chen
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Jingchuan Fan
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yao Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Tingting Wang
- Biochemistry and Molecular Biology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, China
| | - Ziyi Yuan
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Qiang Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Hong Qin
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Jingxin Xu
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
13
|
Walker GE, Merlin S, Zanolini D, Vandoni A, Volpe A, Gaidano G, Valente G, Olivero M, Follenzi A. Factor VIII as a potential player in cancer pathophysiology. J Thromb Haemost 2022; 20:648-660. [PMID: 34847278 PMCID: PMC9306727 DOI: 10.1111/jth.15611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Trousseau sign was the first demonstration of a close relationship between cancer and thrombosis. Currently, venous thromboembolism (VTE) is five to six times more likely to occur in cancer patients, whereas there is a greater risk of cancer diagnoses following thromboses. In considering novel players, factor VIII (FVIII), an essential coagulation cofactor with emerging extracoagulative functions, has been identified as an independent VTE risk factor in cancer; however, the basis of this increase is unknown. OBJECTIVE To investigate the possible direct expression and secretion of FVIII by cancer cells. METHODS Bladder cancer, with a high VTE risk, and normal bladder tissue and epithelium, were used to investigate FVIII. Factor VIII protein and secretion were examined in bladder cancer cell lines. Expanding to other cancers, the Cancer Cell line Encyclopedia database was used to analyze FVIII, tissue factor, FV, FVII, FIX, FX, and von Willebrand factor (VWF) mRNA in 811 cell lines subdivided according to origin. Factor VIII protein synthesis, secretion, and bioactivity were investigated in a profile of cancer cell lines of differing origins. RESULTS AND CONCLUSIONS Although expressed in the normal bladder epithelium, FVIII mRNA and protein were higher in matched bladder neoplasms, with synthesis and secretion of bioactive FVIII evident in bladder cancer cells. This can be extended to other cancer cell lines, with a pattern reflecting the tumor origin, and that is independent of VWF and other relevant players in the coagulation cascade. Here, evidence is provided of a possible independent role for FVIII in cancer-related pathophysiology.
Collapse
Affiliation(s)
- Gillian E. Walker
- Department of Health SciencesUniversità del Piemonte OrientaleNovaraItaly
| | - Simone Merlin
- Department of Health SciencesUniversità del Piemonte OrientaleNovaraItaly
| | - Diego Zanolini
- Department of Health SciencesUniversità del Piemonte OrientaleNovaraItaly
| | - Andrea Vandoni
- Department of Translational MedicineUniversità del Piemonte OrientaleNovaraItaly
| | - Alessandro Volpe
- Department of Translational MedicineUniversità del Piemonte OrientaleNovaraItaly
| | - Gianluca Gaidano
- Department of Translational MedicineUniversità del Piemonte OrientaleNovaraItaly
| | - Guido Valente
- Department of Translational MedicineUniversità del Piemonte OrientaleNovaraItaly
| | - Martina Olivero
- Department of OncologyUniversity of TorinoTorinoItaly
- Candiolo Cancer Institute‐FPOIRCCSCandiolo, TorinoItaly
| | - Antonia Follenzi
- Department of Health SciencesUniversità del Piemonte OrientaleNovaraItaly
| |
Collapse
|
14
|
Yao J, Lang Y, Su H, Dai S, Ying K. Construction of Risk Assessment Model for Venous Thromboembolism After Colorectal Cancer Surgery: A Chinese Single-Center Study. Clin Appl Thromb Hemost 2022; 28:10760296211073748. [PMID: 35167387 PMCID: PMC8851938 DOI: 10.1177/10760296211073748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective A retrospective study was carried out to construct a postoperative venous thromboembolism (VTE) risk assessment model (RAM) applicable for Chinese colorectal cancer patients. Methods 541 Patients who underwent colorectal cancer surgery from June 2019 to May 2020 at Sir-Run-Run-Shaw Hospital affiliated to Zhejiang University School of Medicine were enrolled in this study. Multi-factor analysis was used to determine the independent risk factors of VTE. A novel RAM of VTE which we called Sir-Run-Run-Shaw VTE RAM were constructed basing on the independent risk factors. Another study cohort consisted of 287 colorectal cancer patients underwent surgery from January 2021 to June 2021was used for model evaluation. Results The incidence of VTE after colorectal cancer surgery was 12.0%(65/541). Among the 65 VTE Patients, DVT accounted for 92.3% (60/65) and DVT + PE accounted for 7.7% (5/65). Multi-factor analysis showed that age ≥ 69 years ( P < 0.01), preoperative plasma D-dimer ≥ 0.49 mg/L ( P = .004), stage IV of cancer ( P = .018) and transfusion ( P = .004) are independent risk factors of VTE after surgery. Sir-Run-Run-Shaw VTE RAM includes the above 4 factors, and the total score is 4 points. The score of the low, medium and high risk groups are 0, 1 and ≥2 points. The area under the ROC curve (AUC) of Sir-Run-Run-Shaw VTE RAM is 0.769, while Caprini RAM is 0.656. There is statistical difference between the two risk score tables ( Z = 2.337, P = .0195). Conclusion A VTE RAM is constructed basing on a single center retrospective study. This score table may be applicable for Chinese patients with colorectal cancer surgery.
Collapse
Affiliation(s)
- Jianchang Yao
- Sir-Run-Run-Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Deqing People’s Hospital, Deqing, China
| | - Yina Lang
- Sir-Run-Run-Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Deqing People’s Hospital, Deqing, China
| | - Hua Su
- Sir-Run-Run-Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Dai
- Sir-Run-Run-Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kejing Ying
- Sir-Run-Run-Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Winther-Larsen A, Sandfeld-Paulsen B, Hvas AM. New Insights in Coagulation and Fibrinolysis in Patients with Primary Brain Cancer: A Systematic Review. Semin Thromb Hemost 2021; 48:323-337. [PMID: 34624915 DOI: 10.1055/s-0041-1733961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Patients with primary brain tumors have a high incidence of thrombosis and hemorrhage. The underlying mechanism is believed to be derangement of their hemostatic system. To get nearer a clarification of this, we aimed to systematically review the existing literature regarding primary and secondary hemostasis as well as fibrinolysis in patients with primary brain tumor. The review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The databases PubMed, Embase, and Web of Science were searched on December 15, 2020, without time restrictions. Studies were included if they evaluated at least one blood coagulation and/or fibrinolysis parameter in patients with primary brain cancer. In total, 26 articles including 3,288 patients were included. Overall, increased activity of secondary hemostasis was observed as increased prothrombin fragment 1 + 2 and endogenous thrombin generation levels were found in glioma patients compared with controls. Furthermore, data showed a state of hypofibrinolysis with increased plasminogen activator inhibitor 1 and prolonged clot lysis time in glioma patients. In contrast, no consistent increase in the primary hemostasis was identified; however, data suggested that increased sP-selectin could be a biomarker of increased venous thromboembolism risk and that increased platelet count may be prognostic for survival. Lastly, data indicated that fibrinogen and D-dimer could hold prognostic value. In conclusion, this review indicates that an increased activity of secondary hemostasis and impaired fibrinolysis could be important players in the pathogeneses behind the high risk of thromboembolisms observed in brain cancer patients. Thus, long-term thromboprophylaxis may be beneficial and additional studies addressing this issue are wanted.
Collapse
Affiliation(s)
- Anne Winther-Larsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
16
|
Alterations of the Platelet Proteome in Lung Cancer: Accelerated F13A1 and ER Processing as New Actors in Hypercoagulability. Cancers (Basel) 2021; 13:cancers13092260. [PMID: 34066760 PMCID: PMC8125802 DOI: 10.3390/cancers13092260] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The risk of venous thromboembolism in cancer is nine times higher than in the general population and the second leading cause of death in these patients. Tissue factor and downstream plasmatic coagulation cascade are largely responsible for the risk of thrombosis in cancer. In recent years, it has been increasingly recognised that platelets also play a central role in tumour growth and cancer-associated thrombosis. The underlying molecular mechanisms are largely unknown. In order to comprehensively investigate the biochemical changes in platelets from cancers with high risk of thrombosis, we examined the platelet proteome of brain and lung cancer patients in comparison to sex and age-matched healthy controls. However, we only found alterations in lung cancer, where some of these platelet proteins directly promote thrombosis. One example is the increased amount of the enzyme protein disulfide isomerase, which is clinically investigated as an antithrombotic drug target of the plant-based flavonol quercetin. Abstract In order to comprehensively expose cancer-related biochemical changes, we compared the platelet proteome of two types of cancer with a high risk of thrombosis (22 patients with brain cancer, 19 with lung cancer) to 41 matched healthy controls using unbiased two-dimensional differential in-gel electrophoresis. The examined platelet proteome was unchanged in patients with brain cancer, but considerably affected in lung cancer with 15 significantly altered proteins. Amongst these, the endoplasmic reticulum (ER) proteins calreticulin (CALR), endoplasmic reticulum chaperone BiP (HSPA5) and protein disulfide-isomerase (P4HB) were significantly elevated. Accelerated conversion of the fibrin stabilising factor XIII was detected in platelets of patients with lung cancer by elevated levels of a coagulation factor XIII (F13A1) 55 kDa fragment. A significant correlation of this F13A1 cleavage product with plasma levels of the plasmin–α-2-antiplasmin complex and D-dimer suggests its enhanced degradation by the fibrinolytic system. Protein association network analysis showed that lung cancer-related proteins were involved in platelet degranulation and upregulated ER protein processing. As a possible outcome, plasma FVIII, an immediate end product for ER-mediated glycosylation, correlated significantly with the ER-executing chaperones CALR and HSPA5. These new data on the differential behaviour of platelets in various cancers revealed F13A1 and ER chaperones as potential novel diagnostic and therapeutic targets in lung cancer patients.
Collapse
|
17
|
Lelas A, Greinix HT, Wolff D, Eissner G, Pavletic SZ, Pulanic D. Von Willebrand Factor, Factor VIII, and Other Acute Phase Reactants as Biomarkers of Inflammation and Endothelial Dysfunction in Chronic Graft-Versus-Host Disease. Front Immunol 2021; 12:676756. [PMID: 33995421 PMCID: PMC8119744 DOI: 10.3389/fimmu.2021.676756] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic graft-versus-host disease (cGvHD) is an immune mediated late complication of allogeneic hematopoietic stem cell transplantation (alloHSCT). Discovery of adequate biomarkers could identify high-risk patients and provide an effective pre-emptive intervention or early modification of therapeutic strategy, thus reducing prevalence and severity of the disease among long-term survivors of alloHSCT. Inflammation, endothelial injury, and endothelial dysfunction are involved in cGvHD development. Altered levels of acute phase reactants have shown a strong correlation with the activity of several immune mediated disorders and are routinely used in clinical practice. Since elevated von Willebrand factor (VWF) and factor VIII (FVIII) levels have been described as acute phase reactants that may indicate endothelial dysfunction and inflammation in different settings, including chronic autoimmune diseases, they could serve as potential candidate biomarkers of cGvHD. In this review we focused on reported data regarding VWF and FVIII as well as other markers of inflammation and endothelial dysfunction, evaluating their potential role in cGvHD.
Collapse
Affiliation(s)
- Antonela Lelas
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Daniel Wolff
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Günther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Steven Zivko Pavletic
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Drazen Pulanic
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|