1
|
Haghbin M, Sotoodeh Jahromi A, Hashemi Tayer A, Ghasemi Nejad Z. The Potential Clinical Relevance of Procoagulant Microparticles as Biomarkers of Blood Coagulation in Breast Cancer: A Systematic Review. Asian Pac J Cancer Prev 2025; 26:23-32. [PMID: 39873982 PMCID: PMC12082420 DOI: 10.31557/apjcp.2025.26.1.23] [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: 04/20/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is a global challenge that affects a large portion of individuals, especially women. It has been suggested that microparticles (MPs) can be used as a diagnostic, prognostic, or therapeutic biomarker in various diseases. Moreover, MPs are known to elevate in cancer cases. Platelet-derived MPs (PMPs) play a crucial role in the metastasis of BC, warranting specific focus. This study aimed to explore the involvement of procoagulant MPs in BC. METHODS This systematic review was carried out using the Preferred Reporting Items for Systematic reviews, and Meta-Analyses (PRISMA). Terms defined as MESH keywords were searched PubMed/MEDLINE, Embase, Web of Science, and Cochrane Library searched in from 2011 to March 2024. Experimental and quasi-experimental studies were assessed by the CONSORT checklist. RESULTS Eventually, 15 studies were included. 426 participants were studied in the included articles. The potential clinical relevance of MPs as biomarkers in BC was indicated. Also, the role of MPs in immune modulation and multidrug resistance was approved. PMPs were found to enhance malignant features, including migration and invasion. Moreover, there were lower levels of MPs before neo-adjuvant chemotherapy, suggesting a potential impact of chemotherapy on MPs levels. The study highlights the remarkable capacity of multidrug-resistant BC-derived MPs to alter the phenotype and functionality of immune cells. CONCLUSIONS The findings underscore the intricate interplay between MPs and cellular signaling pathways, shedding light on their potential as diagnostic biomarkers, and therapeutic targets in cancer. Specifically, the association between MPs levels and disease severity, as evidenced by their correlation with tissue-based biomarkers, tumor grading, and distant metastasis, highlights their clinical relevance in prognostication and risk stratification.
Collapse
Affiliation(s)
| | | | - Akbar Hashemi Tayer
- Research Center for Noncommunicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran.
| | | |
Collapse
|
2
|
Su X, Júnior GPDO, Marie A, Gregus M, Figueroa‐Navedo A, Ghiran IC, Ivanov AR. Enhanced proteomic profiling of human plasma-derived extracellular vesicles through charge-based fractionation to advance biomarker discovery potential. J Extracell Vesicles 2024; 13:e70024. [PMID: 39641316 PMCID: PMC11621968 DOI: 10.1002/jev2.70024] [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: 03/03/2024] [Accepted: 11/23/2024] [Indexed: 12/07/2024] Open
Abstract
The study introduces a charge-based fractionation method for fractionating plasma-derived extracellular vesicles (EVs) into sub-populations aimed at the improved purification from free plasma proteins to enhance the diagnostic potential of EV sub-populations for specific pathophysiological states. Here, we present a novel approach for EV fractionation that leverages EVs' inherent surface charges, differentiating them from other plasma components and, thus, reducing the sample complexity and increasing the purity of EVs. The developed method was optimized and thoroughly evaluated using proteomic analysis, transmission electron microscopy, nanoparticle tracking, and western blotting of isolated EVs from healthy donors. Subsequently, we pilot-tested the developed technique for its applicability to real-world specimens using a small set of clinical prostate cancer samples and matched controls. The presented technique demonstrates the effective isolation and fractionation of EV sub-populations based on their surface charge, which may potentially help enhance EV-based diagnostics, biomarker discovery, and basic biology research. The method is designed to be straightforward, scalable, easy-to-use, and it does not require specialized skills or equipment.
Collapse
Affiliation(s)
- Xianyi Su
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Getúlio Pereira de Oliveira Júnior
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Anne‐Lise Marie
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Michal Gregus
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Amanda Figueroa‐Navedo
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| | - Ionita C. Ghiran
- Department of Anesthesia, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Alexander R. Ivanov
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological AnalysisNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
3
|
Mirandola A, Kudriavtsev A, Cofre Muñoz CI, Navarro RC, Macagno M, Daoud S, Sanchez C, Pastor B, Pisareva E, Marin MS, Ruiz JG, Piris A, Rodriguez AG, Gonzalez NS, Vivancos A, Quarà V, Mellano A, Borghi F, Corti G, Marchiò C, Sapino A, Bartolini A, Crisafulli G, Bardelli A, Di Maio M, Lossaint G, Frayssinoux F, Crapez E, Ychou M, Soler RS, Fenocchio E, Fernandez Calotti PX, Mazard T, Vivas CS, Elez E, Di Nicolantonio F, Thierry AR. Post-surgery sequelae unrelated to disease progression and chemotherapy revealed in follow-up of patients with stage III colon cancer. EBioMedicine 2024; 108:105352. [PMID: 39303668 PMCID: PMC11437914 DOI: 10.1016/j.ebiom.2024.105352] [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: 12/07/2023] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND We studied the poorly-known dynamics of circulating DNA (cir-nDNA), as monitored prospectively over an extended post-surgery period, in patients with cancer. METHODS On patients with stage III colon cancer (N = 120), using personalised molecular tags we carried out the prospective, multicenter, blinded cohort study of the post-surgery serial analysis of cir-nDNA concentration. 74 patients were included and 357 plasma samples tested. FINDINGS During post-operative follow-up, the patients' median cir-nDNA concentration was greater (P < 0.0001 in the [43-364 days range]) than both the median value in healthy individuals and the pre-surgery value. These cir-nDNA levels were highly associated with NETs markers (P-value associating MPO and cir-nDNA, and NE and cir-nDNA are 6.6 x 10-17, and 1.9 x 10-7), in accordance with previous reports which indicate that cir-nDNA are NETs by-products. Unexpectedly, in 34 out of 50 patients we found that NETs continued to be formed for an extended duration post-surgery, even in patients without disease progression. Given that this phenomenon was observed in patients without adjuvant CT, and in patients >18 months post-surgery, the data suggest that the persistence of NETs formation is not due to the adjuvant CT. INTERPRETATION (1), Given the inter-patient heterogeneity, the post-surgery cir-nDNA level cannot be considered a reliable value, and caution must be exercised when determining mutation allele frequency or the mutation status; and (2), specific studies must be undertaken to investigate the possible clinical impact of the persistent, low-grade inflammation resulting from elevated NETs levels, such as observed in these post-surgery patients, given that such levels are known to potentially induce adverse cardiovascular or thrombotic events. FUNDING This work was supported by the H2020 European ERA-NET grant on Translational Cancer Research (TRANSCAN-2).
Collapse
Affiliation(s)
- Alexia Mirandola
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Andrei Kudriavtsev
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | | | - Raquel Comas Navarro
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Marco Macagno
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Saidi Daoud
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Cynthia Sanchez
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Brice Pastor
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Ekaterina Pisareva
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Mireia Sanchis Marin
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Javier Gonzalo Ruiz
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Alejandro Piris
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | | | - Nadia Saoudi Gonzalez
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Ana Vivancos
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Virginia Quarà
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Alfredo Mellano
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Felice Borghi
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Giorgio Corti
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Caterina Marchiò
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Medical Sciences, University of Torino, Turin, Italy
| | - Anna Sapino
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Medical Sciences, University of Torino, Turin, Italy
| | - Alice Bartolini
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Giovanni Crisafulli
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology, University of Torino, Turin, Italy
| | - Alberto Bardelli
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology, University of Torino, Turin, Italy
| | | | - Gerald Lossaint
- ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Florence Frayssinoux
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Evelyne Crapez
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Marc Ychou
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Ramon Salazar Soler
- Medical Oncology Department, Institut Català d'Oncologia (ICO) - IDIBELL, Barcelona, Spain
| | - Elisabetta Fenocchio
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | | | - Thibault Mazard
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Cristina Santos Vivas
- Medical Oncology Department, Institut Català d'Oncologia (ICO) - IDIBELL, Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain
| | - Elena Elez
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Federica Di Nicolantonio
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Oncology, University of Torino, Turin, Italy
| | - Alain R Thierry
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France.
| |
Collapse
|
4
|
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
|
5
|
Werner B, Sjoquist KM, Espinoza D, Yip S, Chang G, Cummins MM, Mileshkin L, Ananda S, Shannon C, Friedlander M, Warton K, Ford CE. Cell-free DNA in plasma and ascites as a biomarker of bevacizumab response- a translational research sub-study of the REZOLVE (ANZGOG-1101) clinical trial. Transl Oncol 2024; 43:101914. [PMID: 38417292 DOI: 10.1016/j.tranon.2024.101914] [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: 07/10/2023] [Revised: 11/30/2023] [Accepted: 02/13/2024] [Indexed: 03/01/2024] Open
Abstract
OBJECTIVE To investigate cell-free DNA (cfDNA) in plasma and ascites and its association with clinical outcomes (paracentesis-free interval, overall survival) and CA125 level in participants with advanced ovarian cancer, treated with palliative intraperitoneal bevacizumab to delay re-accumulation of ascites. METHODS cfDNA was extracted from 0.3 to 1 mL samples from 20/24 participants of the REZOLVE trial. Standard and methylation-specific PCRs were performed to measure 3 biomarkers: total cfDNA (Alu), tumour-derived cfDNA (ctDNA, methylated IFFO1 promoter) and endothelium-derived cfDNA (ec-cfDNA, unmethylated CDH5 promoter). Values were correlated to clinical outcomes. RESULTS cfDNA was detected in all samples, with higher yield in ascites (mean 669 ng/mL) than plasma (mean 75 ng/mL, p < 0.0001). Ascites had a higher ctDNA proportion than plasma (74 % vs. 20 %, p < 0.0001) and plasma had a higher ec-cfDNA proportion than ascites (24 % vs. 16 %, p < 0.002). High ctDNA proportion (>75 %) in ascites was associated with a significantly shorter paracentesis-free interval (median interval 47.5 versus 84 days, hazard ratio (HR) 2.21, 95 % confidence interval (CI) 0.85 to 5.73, p = 0.039) and ctDNA presence in plasma was unfavourable for survival (median survival 56 versus 242 days, HR 3.21, 95 % CI 1.15 to 9.00, p = 0.008). A significant positive correlation was observed between ctDNA proportion in plasma and CA125 level (p = 0.012). No significant difference in total cfDNA, ctDNA nor ec-cfDNA was observed between participants who were responders versus non-responders. CONCLUSION Sufficient cfDNA was detected in both plasma and ascites to study three biomarkers. These samples can provide useful information and should be considered in the design of future ovarian cancer trials.
Collapse
Affiliation(s)
- Bonnita Werner
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Katrin M Sjoquist
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - David Espinoza
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Sonia Yip
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Garry Chang
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Michelle M Cummins
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | | | - Sumitra Ananda
- Peter MacCallum Cancer Centre, Melbourne, Australia; Western Health, Furlong Road, St Albans, Australia; Epworth Freemasons Hospital, East Melbourne, Australia; Department of Medicine, Western Health, University of Melbourne, Melbourne, Australia
| | | | - Michael Friedlander
- Prince of Wales Hospital, Sydney, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Kristina Warton
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Caroline E Ford
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia.
| |
Collapse
|
6
|
Al Harrasi S, Al Salmi I, Al Busaidi N. Acute Pulmonary Embolism in Oman: A Situational Analysis. Oman Med J 2023; 38:e574. [PMID: 38343447 PMCID: PMC10858989 DOI: 10.5001/omj.2023.125] [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: 02/27/2023] [Accepted: 07/15/2023] [Indexed: 12/16/2024] Open
Abstract
OBJECTIVES Pulmonary embolism (PE) is increasingly prevalent in Oman. The aim of this situational analysis of PE among Omani patients was to study its predisposing factors, diagnostic issues, and optimal management. METHODS In this retrospective cross-sectional situational analysis, the subjects were Omani patients who were diagnosed with acute PE using computed tomography pulmonary angiogram from 2010-2021. The required data was collected from the hospital database and statistically analyzed. RESULTS The subjects were 438 patients diagnosed with PE, with a mean age of 53.3±18.5 years and mean body mass index of 29.7±7.3 kg/m2. Males were in a slight majority (223; 50.9%) and were older (55.0±18.5 years) than females (51.5±18.3 years). Two-thirds of the PE patients had hypertension and one-third had diabetes mellitus. Most patients presented with dyspnea and chest pain. Syncope was more common in females compared to males who mostly presented with hemoptysis. Nearly half of the patients had abnormal echocardiogram, and males had lower mean ejection fraction compared to females. Different modalities of management were used to treat patients' PE. Heparin was the most used anticoagulant followed by warfarin and direct oral anticoagulants. Out of the 122 PE patients who died during the study period, PE was the direct cause of death of 68 patients. The mortality was higher in males than in females. CONCLUSIONS The incidence of acute PE was similar among both male and female Omani patients. However, there were sex differences in risk factors, symptoms, investigations, and management of PE. Females presented with more severe PE, but mortality was higher in males.
Collapse
Affiliation(s)
- Sally Al Harrasi
- Internal Medicine Residency Training Program, Oman Medical Specialty Board, Muscat, Oman
| | - Issa Al Salmi
- Internal Medicine Residency Training Program, Oman Medical Specialty Board, Muscat, Oman
- Department of Renal Medicine, Royal Hospital, Muscat, Oman
| | | |
Collapse
|
7
|
Ha H, Ko YH, Kim K, Hong J, Lee GW, Jeong SH, Bang SM, Yoon SS. Application of the Khorana score for cancer-associated thrombosis prediction in patients of East Asian ethnicity undergoing ambulatory chemotherapy. Thromb J 2023; 21:63. [PMID: 37271814 DOI: 10.1186/s12959-023-00505-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND The Khorana score (KS) has not been well studied in East Asian cancer patients, who have different genetic backgrounds for inherited thrombophilia, body metabolism, and cancer epidemiology. METHODS By using the Common Data Model, we retrospectively collected deidentified data from 11,714 consecutive newly diagnosed cancer patients who underwent first-line chemotherapy from December 2015 to December 2021 at a single institution in Korea, and we applied the KS for cancer-associated thrombosis (CAT) prediction. Age at diagnosis, sex, and use of highly thrombogenic chemotherapeutics were additionally investigated as potential risk factors for CAT development. RESULTS By 6 months after chemotherapy initiation, 207 patients (1.77%) experienced CAT. Only 0.4% had a body mass index (BMI) ≥ 35 kg/m2 and changing the cutoff to 25 kg/m2 improved the prediction of CAT. Age ≥ 65 years and the use of highly thrombogenic chemotherapeutics were independently associated with CAT development. KS values of 1 ~ 2 and ≥ 3 accounted for 52.3% and 7.6% of all patients, respectively, and the incidence of CAT in these groups was 2.16% and 4.16%, respectively, suggesting a lower incidence of CAT in the study population than in Westerners. The KS component regarding the site of cancer showed a good association with CAT development but needed some improvement. CONCLUSION The KS was partially validated to predict CAT in Korean cancer patients undergoing modern chemotherapy. Modifying the BMI cutoff, adding other risk variables, and refining the use of cancer-site data for CAT risk prediction may improve the performance of the KS for CAT prediction in East Asian patients.
Collapse
Affiliation(s)
- Hyerim Ha
- Department of Internal Medicine, Inha University College of Medicine, Inha University Hospital, Incheon, Republic of Korea
| | - Yeh-Hee Ko
- Department of Applied Statistics, Yonsei University, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine & Advanced Technology, Department of Medicine, College of Medicine, Seoul National University Hospital, Seoul National University, 101 Daehak-ro, Jongno-gu, Adjunct, Seoul, Republic of Korea.
- Department of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Junshik Hong
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Gyeong-Won Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Seong Hyun Jeong
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Soo-Mee Bang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National Bundang Hospital, Seongnam, Republic of Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
8
|
Ben S, Huang X, Shi Y, Xu Z, Xiao H. Change in cytokine profiles released by mast cells mediated by lung cancer-derived exosome activation may contribute to cancer-associated coagulation disorders. Cell Commun Signal 2023; 21:97. [PMID: 37143160 PMCID: PMC10161433 DOI: 10.1186/s12964-023-01110-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/22/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Coagulation disorders are a significant cause of lung cancer mortality. Although mast cells are known to play a role in coagulation abnormalities, their specific role in this process has not yet been elucidated. METHOD We detected mast cells in the tumor microenvironment using single-cell sequencing data and examined their correlation with thrombosis-related genes, neutrophil-related genes, neutrophil extracellular trap-related signature genes, and immune infiltration levels in lung cancer patients through bioinformatics analysis. Bone marrow mast cell uptake of exosomes isolated from the lung adenocarcinoma cell line A549, which were labeled using PKH67, was observed using confocal microscopy. Mast cell degranulation was detected by measuring the β-hexosaminidase release rate. Additionally, cytokine array analysis was performed to identify altered mediators released by bone marrow mast cells after uptake of the exosomes. RESULTS In our study, we found a close correlation between the proportion of mast cells in lung cancer patients and the expression levels of thrombosis-related genes and neutrophil extracellular trap signature genes, both of which play a key role in thrombophilic disorder. Moreover, we discovered that lung cancer cell-derived exosomes can be taken up by mast cells, which in turn become activated to release procoagulant mediators. CONCLUSION Our study shows that exosomes derived from lung cancer cells can activate mast cells to release procoagulants that may contribute to abnormal blood clotting in lung cancer patients. Video Abstract.
Collapse
Affiliation(s)
- Suqin Ben
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiulin Huang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxin Shi
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziheng Xu
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
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: 9] [Impact Index Per Article: 4.5] [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
|
10
|
Cohen A, Noxon V, Dhamane A, Bruette R, Shah S, Hines DM, Alfred T, Luo X. Effectiveness and safety of anticoagulants among venous thromboembolism cancer patients with and without brain cancer. Thromb Res 2023; 226:117-126. [PMID: 37146438 DOI: 10.1016/j.thromres.2023.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 05/07/2023]
Abstract
INTRODUCTION Patients with brain cancer are at a high risk of developing venous thromboembolism (VTE) and are underrepresented in clinical trials. This study compared the risk of recurrent VTE (rVTE), major bleeding (MB), and clinically relevant non-major bleeding (CRNMB) among VTE cancer patients initiating apixaban, low molecular weight heparin (LMWH), or warfarin stratified by patients with brain vs other cancer types. MATERIALS AND METHODS Active cancer patients initiating apixaban, LMWH, or warfarin within 30 days after VTE diagnosis were identified from 4 US commercial and the Medicare databases. Inverse probability of treatment weights (IPTW) was used to balance patient characteristics. Cox proportional hazards models were used to evaluate the interaction between brain cancer status and treatment on outcomes (rVTE, MB, and CRNMB), with a p-value <0.1 indicating a significant interaction. RESULTS Of 30,586 patients with active cancer (5 % had brain cancer), apixaban (vs. LMWH and warfarin) was associated with lower risk of rVTE, MB, and CRNMB. Generally, no significant interactions (P > 0.1) were found between brain cancer status and anticoagulant treatment across outcomes. The exception was MB for apixaban [vs LMWH (p-value for interaction = 0.091)] with a higher reduction among those with brain cancer (HR = 0.32) than those with (HR = 0.72) other cancer. CONCLUSIONS Among VTE patients with all types of cancer, apixaban (vs LMWH and warfarin) was associated with a lower risk of rVTE, MB, and CRNMB. In general, anticoagulant treatment effects were not significantly different between VTE patients with brain cancer and those with other cancer.
Collapse
Affiliation(s)
- Alexander Cohen
- Department of Hematological Medicine, Guy's & St Thomas' NHS Foundation Trust, King's College London, Westminster Bridge Road, London, UK.
| | | | - Amol Dhamane
- Bristol Myers Squibb Company, Lawrenceville, NJ, USA
| | | | | | | | | | | |
Collapse
|
11
|
Mackman N, Hisada Y. Circulating Tumor Cells and Cancer-Associated Venous Thrombosis: A Missing Link. Arterioscler Thromb Vasc Biol 2023; 43:160-162. [PMID: 36453278 PMCID: PMC10234083 DOI: 10.1161/atvbaha.122.318715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Affiliation(s)
- Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
12
|
Liu Y, Gao L, Fan Y, Ma R, An Y, Chen G, Xie Y. Discovery of protein biomarkers for venous thromboembolism in non-small cell lung cancer patients through data-independent acquisition mass spectrometry. Front Oncol 2023; 13:1079719. [PMID: 36874092 PMCID: PMC9976579 DOI: 10.3389/fonc.2023.1079719] [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: 10/25/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Objective Non-small cell lung cancer (NSCLC) patients present a high incidence of venous thromboembolism (VTE) with poor prognosis. It is crucial to identify and diagnose VTE early. The study aimed to identify potential protein biomarkers and mechanism of VTE in NSCLC patients via proteomics research. Methods Proteomic analysis of the human plasma was performed through data-independent acquisition mass spectrometry for 20 NSCLC patients with VTE, and 15 NSCLC patients without VTE. Significantly differentially expressed proteins were analyzed by multiple bioinformatics method for further biomarker analysis. Results A total of 280 differentially expressed proteins were identified in VTE and non-VTE patients, where 42 were upregulated and 238 were downregulated. These proteins were involved in acute-phase response, cytokine production, neutrophil migration and other biological processes related to VTE and inflammation. Five proteins including SAA1, S100A8, LBP, HP and LDHB had significant change between VTE and non-VTE patients, with the area under the curve (AUC) were 0.8067, 0.8308, 0.7767, 0.8021, 0.8533, respectively. Conclusions SAA1, S100A8, LBP, HP and LDHB may serve as potential plasma biomarkers for diagnosis VTE in NSCLC patients.
Collapse
Affiliation(s)
- Yanhong Liu
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Lan Gao
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Yanru Fan
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Rufei Ma
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Yunxia An
- Department of Respiratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, China
| | - Guanghui Chen
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Yan Xie
- Department of Laboratory Medcine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, and People's Hospital of Henan University, Zhengzhou, Henan, China
| |
Collapse
|
13
|
Lee LH, Danchaivijitr P, Uaprasert N, Gill H, Sacdalan DL, Ho GF, Parakh R, Pai P, Lee JK, Rey N, Cohen AT. Safe and effective treatment of venous Thromboembolism associated with Cancer: focus on direct Oral Anticoagulants in Asian patients. Exp Hematol Oncol 2022; 11:79. [PMID: 36303259 PMCID: PMC9615183 DOI: 10.1186/s40164-022-00331-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022] Open
Abstract
Cancer-associated thrombosis (CAT) poses a significant disease burden and the incidence in Asian populations is increasing. Anticoagulation is the cornerstone of treatment, but can be challenging due to the high bleeding risk in some cancers and the high risk of recurrent venous thromboembolism (VTE) in patients with malignancies. Direct oral anticoagulants (DOACs) are well established as first-choice treatments for VTE in non-cancer patients, offering a more convenient and less invasive treatment option than low-molecular-weight heparin (LMWH). Asian patients have exhibited comparable efficacy and safety outcomes with other races in trials of DOACs for VTE in the general population. Although no specific data are available in Asian patients with CAT, results from randomized controlled trials of apixaban, edoxaban, or rivaroxaban versus the LMWH, dalteparin, indicate that DOACs are a reasonable alternative to LMWH for anticoagulation in Asian patients with CAT. This is further supported by analyses of real-world data in Asian populations demonstrating the efficacy and safety of DOACs in Asian patients with CAT. Apixaban, edoxaban, or rivaroxaban are recommended in the most recently updated international guidelines as first-line therapy for CAT in patients without gastrointestinal or genitourinary cancers and at low risk of bleeding. An increased risk of major gastrointestinal bleeding was evident with edoxaban or rivaroxaban, but not apixaban, versus dalteparin in the clinical trials, suggesting that apixaban could be a safe alternative to LMWH in patients with gastrointestinal malignancies. Determining the optimal anticoagulant therapy for patients with CAT requires careful consideration of bleeding risk, tumor type, renal function, drug-drug interactions, financial costs, and patients' needs and preferences.
Collapse
Affiliation(s)
- Lai Heng Lee
- Haematology Department, Singapore General Hospital, Bukit Merah, Singapore
| | - Pongwut Danchaivijitr
- Division of Medical Oncology, Department of Internal Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Noppacharn Uaprasert
- Research Unit in Translational Hematology, Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Harinder Gill
- Department of Medicine, The University of Hong Kong, 102 Pokfulam Road, Pokfulam Hong Kong, Pokfulam, Hong Kong, China.
| | | | - Gwo Fuang Ho
- Clinical Oncology, University Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Rajiv Parakh
- Division of Peripheral Vascular & Endovascular Service, Medanta-Medicity, Gurgaon, Haryana, India
| | - Paresh Pai
- Vascular and Endovascular Surgery, The Vascular Clinic, Mumbai, India
| | - Jen-Kuang Lee
- Cardiology Department, National Taiwan University Hospital, Taipei, Taiwan
| | - Nannette Rey
- de La Salle Medical and Health Sciences Institute, Dasmarinas Cavite, Philippines
| | - Alexander T Cohen
- Department of Haematological Medicine, Guy's and St Thomas' Hospitals, NHS Trust, King's College, London, UK
| |
Collapse
|
14
|
Li X, Lu D, Zhang Z, Zhang Y, Wang J, Hu Y. Prognostic value of plasma D-dimer levels in advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors: a retrospective study. J Thorac Dis 2022; 14:4125-4135. [PMID: 36389301 PMCID: PMC9641356 DOI: 10.21037/jtd-22-1363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/19/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Plasma D-dimer is of great significance for the clinical exclusion of tumor-related thrombosis. Previous studies have shown its predictive role in non-small cell lung cancer (NSCLC) treated with chemotherapy. However, whether pretreatment D-dimer could predict the efficacy and prognosis in NSCLC patients treated with immune checkpoint inhibitors (ICIs) remains unclear. METHODS Advanced NSCLC patients treated with ICIs at the Chinese PLA General Hospital between January 2015 and March 2019 were enrolled. Patients were divided into a pretreatment normal D-dimer group (≤0.5 µg/mL) and high D-dimer group (>0.5 µg/mL). Optimization-based approach was applied to balance baseline covariates between the 2 groups, including age, sex, histological type, smoking history, stage, Eastern Cooperative Oncology Group Performance Status (ECOG PS), lines of treatment, ICI drugs, brain metastasis, treatment type, and D-dimer levels. Kaplan-Meier analysis and Cox proportional hazards model were used for analyzing survival data, including progression-free survival (PFS, the time from initial ICI treatment to PD or death), overall survival (OS, the time between initial ICI treatment and death), and hazard ratio (HR). Follow-up of all patients was performed by searching electronic medical records and counseling telephone. The follow-up cut-off date was July 6, 2020. RESULTS This study included 277 advanced NSCLC patients. Among the enrolled patients, 23.1% were female, 64.6% had non-squamous cell lung cancer, and 79.4% were stage IV. Univariate and multivariate analysis showed that pretreatment high D-dimer levels were independently associated with shortened PFS and OS (P<0.01). Subgroup analysis confirmed that pretreatment high D-dimer levels were associated with poor prognosis in most subsets. After balancing baseline covariates between the high D-dimer group and normal D-dimer group, the results indicated that patients with pretreatment high D-dimer levels had significantly shorter PFS [median: 6.4 vs. 11.5 months; HR, 1.70; 95% confidence ratio (CI): 1.25-2.37; P<0.001] and OS (median: 12.7 vs. 30.4 months; HR, 2.29; 95% CI: 1.54-3.41; P<0.001) than those with pretreatment normal D-dimer levels. CONCLUSIONS Pretreatment plasma D-dimer could serve as a convenient prognostic biomarker for advanced NSCLC patients receiving ICI treatment. Patients with pretreatment high D-dimer levels may have poor PFS and OS.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Zhibo Zhang
- Department of Cardiothoracic Surgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Yuning Zhang
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Jinliang Wang
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yi Hu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Del Vecchio S, Terlizzi C, Pellegrino S, Altobelli GG, Fonti R. What molecular imaging of cancer patients can teach us about COVID-19. EUROPEAN PHYSICAL JOURNAL PLUS 2022; 137:1069. [PMID: 36158866 PMCID: PMC9484336 DOI: 10.1140/epjp/s13360-022-03262-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/02/2022] [Indexed: 06/16/2023]
Abstract
COVID-19 pandemic had a great impact on health systems and cancer care worldwide. Patients with cancer who develop COVID-19 are at high risk of severe outcomes and clarifying the determinants of such vulnerability of cancer patients would be of great clinical benefit. While the mechanisms of SARS-CoV-2 infection have been elucidated, the pathogenetic pathways leading to severe manifestations of the disease are largely unknown. Critical manifestations of COVID-19 mainly occur in elderly patients and in patients with serious comorbidities including cancer. Efforts to understand the intersection of pathways between severe manifestations of COVID-19 and cancer may shed light on the pathogenesis of critical illness in COVID-19 patients. Here, we will focus our attention on two major fields of potential intersection between COVID-19 and cancer, namely the dysfunction of immune system and the prothrombotic state that can occur in both COVID-19 and cancer patients, testing whether cancer imaging can provide clues to better understand such interactions.
Collapse
Affiliation(s)
- Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Cristina Terlizzi
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Sara Pellegrino
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Giovanna G. Altobelli
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
16
|
Zhou Y, Xu Z, Liu Z. Impact of Neutrophil Extracellular Traps on Thrombosis Formation: New Findings and Future Perspective. Front Cell Infect Microbiol 2022; 12:910908. [PMID: 35711663 PMCID: PMC9195303 DOI: 10.3389/fcimb.2022.910908] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022] Open
Abstract
Thrombotic diseases seriously endanger human health, neutrophils and neutrophil extracellular traps (NETs) play an important role in abnormal thrombus formation. NETs are extracellular structures released by neutrophils upon stimulation by pathogens. NETs include neutrophil elastase (NE), myeloperoxidase (MPO), cathepsin G and other active substances. The network structure provided by NETs can prevent the spread of pathogens and effectively kill and eliminate pathogens. However, the components of NETs can also abnormally activate the coagulation pathway and participate in the formation of pathological thrombi. This review aims to summarize the mechanisms of NETs formation in detail; the research progress of NETs in venous thrombosis, arterial thrombosis, acquired disease-associated thrombosis, sepsis coagulation disorder; as well as the strategies to target NETs in thrombosis prevention and treatment.
Collapse
Affiliation(s)
| | - Zhendong Xu
- *Correspondence: Zhiqiang Liu, ; Zhendong Xu,
| | | |
Collapse
|
17
|
Campello E, Bosh F, Simion C, Spiezia L, Simioni P. Mechanisms of thrombosis in pancreatic ductal adenocarcinoma. Best Pract Res Clin Haematol 2022; 35:101346. [DOI: 10.1016/j.beha.2022.101346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022]
|
18
|
Beck S, Hochreiter B, Schmid JA. Extracellular Vesicles Linking Inflammation, Cancer and Thrombotic Risks. Front Cell Dev Biol 2022; 10:859863. [PMID: 35372327 PMCID: PMC8970602 DOI: 10.3389/fcell.2022.859863] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) being defined as lipid-bilayer encircled particles are released by almost all known mammalian cell types and represent a heterogenous set of cell fragments that are found in the blood circulation and all other known body fluids. The current nomenclature distinguishes mainly three forms: microvesicles, which are formed by budding from the plasma membrane; exosomes, which are released, when endosomes with intraluminal vesicles fuse with the plasma membrane; and apoptotic bodies representing fragments of apoptotic cells. Their importance for a great variety of biological processes became increasingly evident in the last decade when it was discovered that they contribute to intercellular communication by transferring nucleotides and proteins to recipient cells. In this review, we delineate several aspects of their isolation, purification, and analysis; and discuss some pitfalls that have to be considered therein. Further on, we describe various cellular sources of EVs and explain with different examples, how they link cancer and inflammatory conditions with thrombotic processes. In particular, we elaborate on the roles of EVs in cancer-associated thrombosis and COVID-19, representing two important paradigms, where local pathological processes have systemic effects in the whole organism at least in part via EVs. Finally, we also discuss possible developments of the field in the future and how EVs might be used as biomarkers for diagnosis, and as vehicles for therapeutics.
Collapse
Affiliation(s)
- Sarah Beck
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| | - Bernhard Hochreiter
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Johannes A. Schmid
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| |
Collapse
|
19
|
Daguenet E, Maison M, Tinquaut F, Giroux EA, Bertoletti L, Suchaud JP, Rancoule C, Guy JB, Magné N. Venous thromboembolism and radiation therapy: The final radiation-induced thrombosis study analysis. Cancer Med 2022; 11:1753-1762. [PMID: 35199492 PMCID: PMC9041075 DOI: 10.1002/cam4.4559] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/10/2021] [Accepted: 01/07/2022] [Indexed: 12/21/2022] Open
Abstract
Background Thromboembolic events frequently complicate the course of malignancy and represent a major cause of morbidity and mortality in cancer patients. In contrast to chemotherapy and other systemic therapies, little is known about the impact of ionizing radiations on the incidence of venous thromboembolism (VTE) in cancer patients. Methods In the present prospective study, we aimed to investigate the incidence, management, and outcome of VTE in newly diagnosed cancer patients who received curative radiotherapy. Results VTE was found in 8 patients, out of 401 patients at a median time of 80 days after radiotherapy initiation. The incidence rate of VTE at 6 months post‐treatment was 2% (95% CI, 0.9–3.7), with 50% of cases occurring during the radiotherapy course and 50% of cases in patients who received or were receiving chemotherapy. As none of the patients harbored a personal history of VTE, no prophylactic measure was initiated during cancer therapy. Most patients received monotherapy with low‐molecular‐weight heparin and were still on surveillance at the end of the study. No specific clinical risk factor was identified that might systematically indicate the need of thromboprophylaxis in the context of curative radiotherapy. Conclusions Although this pan‐cancer descriptive study did not relate an increased risk of short‐term thrombosis following ionizing radiation, it provides important insight as a basis for future studies with subcategories of cancer, in order to in fine guide further recommendations in frail patients. Clinical trial registration number NCT02696447.
Collapse
Affiliation(s)
- Elisabeth Daguenet
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France.,Département de Radiothérapie, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Mathilde Maison
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France.,Département de Radiothérapie, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Fabien Tinquaut
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Eric-Alban Giroux
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Laurent Bertoletti
- Service de Médecine Vasculaire et Thérapeutique, CHU de St-Etienne, Saint-Etienne, France.,INSERM, UMR1059, Equipe Dysfonction Vasculaire et Hémostase, Université Jean-Monnet, Saint-Etienne, France.,INSERM, Saint-Etienne, France
| | | | - Chloé Rancoule
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France.,Département de Radiothérapie, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Jean-Baptiste Guy
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| | - Nicolas Magné
- Département Universitaire de la Recherche et de l'Enseignement, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France.,Département de Radiothérapie, Institut de Cancérologie Lucien Neuwirth, St-Priest-en-Jarez, France
| |
Collapse
|
20
|
Ciepiela O, Małecka-Giełdowska M, Czyżewska E. Neutrophil Extracellular Traps (NETs) and Hypercoagulability in Plasma Cell Dyscrasias-Is This Phenomenon Worthy of Exploration? J Clin Med 2021; 10:jcm10225243. [PMID: 34830525 PMCID: PMC8624472 DOI: 10.3390/jcm10225243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Plasma cell dyscrasias (PCDs) are neoplastic diseases derived from plasma cells. Patients suffering from PCDs are at high risk of hypercoagulability and thrombosis. These conditions are associated with disease-related factors, patient-related factors, or the use of immunomodulatory drugs. As PCDs belong to neoplastic diseases, some other factors related to the cancer-associated hypercoagulability state in the course of PCDs are also considered. One of the weakest issues studied in PCDs is the procoagulant activity of neutrophil extracellular traps (NETs). NETs are web-like structures released from neutrophils in response to different stimuli. These structures are made of deoxyribonucleic acid (DNA) and bactericidal proteins, such as histones, myeloperoxidase, neutrophil elastase, and over 300 other proteins, which are primarily stored in neutrophil granules. NETs immobilize, inactivate the pathogens, and expose them to specialized cells of immune response. Despite their pivotal role in innate immunity, they contribute to the development and exacerbation of autoimmune diseases, trigger inflammatory response, or even facilitate the formation of cancer metastases. NETs were also found to induce activity of coagulation and are considered one of the most important factors inducing thrombosis. Here, we summarize how PCDs influence the release of NETs, and hypothesize whether NETs contribute to hypercoagulability in PCDs patients.
Collapse
|
21
|
Kobayashi S, Koizume S, Takahashi T, Ueno M, Oishi R, Nagashima S, Sano Y, Fukushima T, Tezuka S, Morimoto M, Nakamura S, Narimatsu H, Ruf W, Miyagi Y. Tissue factor and its procoagulant activity on cancer-associated thromboembolism in pancreatic cancer. Cancer Sci 2021; 112:4679-4691. [PMID: 34382298 PMCID: PMC8586686 DOI: 10.1111/cas.15106] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/09/2021] [Accepted: 08/09/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer frequently involves cancer-associated thromboembolism, which is strongly associated with poor prognosis. Tissue factor, a blood coagulation factor largely produced in cancer patients as a component of extracellular vesicles, plays a key role in the incidence of cancer-associated thromboembolism in patients with pancreatic cancer. However, no prospective studies have been published on the relationship between tissue factor and cancer-associated thromboembolism or patient clinical characteristics, including recent chemotherapy regimens. Thus, we aimed to address this in a Japanese cohort of 197 patients and 41 healthy volunteers. Plasma tissue factor levels were measured by ELISAs preevaluated by tissue factor specificity. Multivariable analysis was used to identify independent predictors of cancer-associated thromboembolism. We found that the cancer-associated thromboembolism rate in the patient cohort was 6.6% (4.6%, venous thromboembolism; 2.0%, arterial thromboembolism). Tissue factor levels of 100 pg/mL or higher at patient registration were predictive of cancer-associated thromboembolism, with positive and negative predictive values of 23.1% and 94.6%, respectively. Multivariable analysis showed that plasma tissue factor levels were an independent predictive factor for cancer-associated thromboembolism, with a risk ratio of 5.54 (95% confidence interval, 1.02-30.09). Unlike in healthy volunteers and patients without cancer-associated thromboembolism, tissue factor levels were highly correlated with extracellular vesicles' procoagulant activity in patients developing cancer-associated thromboembolism. Taken together, our data show that the tissue factor levels at patient registration were a predictive factor for cancer-associated thromboembolism in this cohort of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Satoshi Kobayashi
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Tomoko Takahashi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Ritsuko Oishi
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shuhei Nagashima
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yusuke Sano
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shun Tezuka
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Manabu Morimoto
- Department of Gastroenterology, Hepatobiliary and Pancreatic Medical Oncology Division, Kanagawa Cancer Center, Kanagawa, Japan
| | - Sho Nakamura
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Hiroto Narimatsu
- Cancer Prevention and Control Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| |
Collapse
|
22
|
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
|
23
|
Pedersen S, Kristensen AF, Falkmer U, Christiansen G, Kristensen SR. Increased activity of procoagulant factors in patients with small cell lung cancer. PLoS One 2021; 16:e0253613. [PMID: 34288927 PMCID: PMC8294523 DOI: 10.1371/journal.pone.0253613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022] Open
Abstract
Small cell lung cancer (SCLC) patients have augmented risk of developing venous thromboembolism, but the mechanisms triggering this burden on the coagulation system remain to be understood. Recently, cell-derived microparticles carrying procoagulant phospholipids (PPL) and tissue factor (TF) in their membrane have attracted attention as possible contributors to the thrombogenic processes in cancers. The aims of this study were to assess the coagulation activity of platelet-poor plasma from 38 SCLC patients and to provide a detailed procoagulant profiling of small and large extracellular vesicles (EVs) isolated from these patients at the time of diagnosis, during and after treatment compared to 20 healthy controls. Hypercoagulability testing was performed by thrombin generation (TG), procoagulant phospholipid (PPL), TF activity, Protein C, FVIII activity and cell-free deoxyribonucleic acid (cfDNA), a surrogate measure for neutrophil extracellular traps (NETs). Our results revealed a coagulation activity that is significantly increased in the plasma of SCLC patients when compared to age-related healthy controls, but no substantial changes in coagulation activity during treatment and at follow-up. Although EVs in the patients revealed an increased PPL and TF activity compared with the controls, the TG profiles of EVs added to a standard plasma were similar for patients and controls. Finally, we found no differences in the coagulation profile of patients who developed VTE to those who did not, i.e. the tests could not predict VTE. In conclusion, we found that SCLC patients display an overall increased coagulation activity at time of diagnosis and during the disease, which may contribute to their higher risk of VTE.
Collapse
Affiliation(s)
- Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- * E-mail:
| | - Anne Flou Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ursula Falkmer
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Søren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
24
|
Gazzeri R, Telera S, Galarza M, Callovini GM, Sperduti I, Alfieri A. Surgical treatment of solitary intradural extramedullary spinal cord metastases from solid cancers of non-neurogenic origin. A multicenter study. J Neurooncol 2021; 154:101-112. [PMID: 34255272 DOI: 10.1007/s11060-021-03804-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/02/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Intradural extramedullary spinal metastases (IESM) represent an extremely rare manifestation of systemic cancer. We evaluated the surgical indications, complications and outcome in a series of 43 patients with solitary intradural extramedullary metastases originating from solid cancer of non-neurogenic origin. METHODS Patients' age, histopathological diagnoses of primary cancer, tumor size, spinal location, and extramedullary tumor dissemination were collected. Preoperative functional status, pre- and post-operative neurological status, extent of the tumor resection were also analyzed. RESULTS The majority of IEMS occurred in the thoracic area, with the most common presenting symptoms ranging from motor (76.7%) to sensory (72%) deficits. Gross total resection was achieved in 55.8% of cases, while In 44.2% of patients a subtotal resection was performed due to strong adherence between the tumor and neural tissue. After surgery, 72.1% of patients exhibited improvement of symptoms in terms of pain relief and partial recovery of motor and/or sensory deficits, while neurologic functional status was severely affected postoperatively in 3 patients. CONCLUSION Although there was no statistical significance between the different parameters and overall survival, KPS and the presence of other metastases were the strongest prognostic factors for overall survival and postoperative neurologic outcome.
Collapse
Affiliation(s)
- Roberto Gazzeri
- Department of Neurosurgery, IRCCS Istituto Nazionale Tumori "Regina Elena", Rome, Italy.
- Department of Pain Therapy, San Giovanni-Addolorata Hospital, Rome, Italy.
| | - Stefano Telera
- Department of Neurosurgery, IRCCS Istituto Nazionale Tumori "Regina Elena", Rome, Italy
| | - Marcelo Galarza
- Regional Service of Neurosurgery, "Virgen de La Arrixaca" University Hospital, Murcia, Spain
| | | | - Isabella Sperduti
- Department of Biostatistics, IRCCS Istituto Nazionale Tumori "Regina Elena", Rome, Italy
| | - Alex Alfieri
- Cantonal Hospital Winterthur, Winterthur, Switzerland
- Neurosurgery, Faculty of Health Sciences, The Brandenburg Medical School Theodor Fontane, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The University of Potsdam, Potsdam, Germany
| |
Collapse
|
25
|
Skille H, Paulsen B, Hveem K, Severinsen MT, Gabrielsen ME, Kristensen SR, Næss IA, Hindberg K, Tjønneland A, Brækkan SK, Hansen JB. Prothrombotic genotypes and risk of venous thromboembolism in occult cancer. Thromb Res 2021; 205:17-23. [PMID: 34237679 DOI: 10.1016/j.thromres.2021.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Studies have reported that the combination of some prothrombotic genotypes and overt cancer yields a synergistic effect on VTE risk. Whether individual prothrombotic genotypes or number of risk alleles in a genetic risk score (GRS) affect VTE risk in occult cancer have not been addressed. The aim of this study was to investigate the joint effect of five prothrombotic genotypes and occult cancer on VTE risk. METHODS Cases with incident VTE (n = 1566) and a subcohort (n = 14,537) were sampled from the Scandinavian Thrombosis and Cancer Cohort (1993-2012). Five single nucleotide polymorphisms previously reported in a GRS were genotyped: ABO (rs8176719), F5 (rs6025), F2 (rs1799963), FGG (rs2066865) and F11 (rs2036914). Hazard ratios (HRs) for VTE by individual SNPs and GRS were estimated according to non-cancer and occult cancer (one year preceding a cancer diagnosis) exposure. RESULTS Occult cancer occurred in 1817 subjects, and of these, 93 experienced a VTE. The VTE risk was 4-fold higher (HR 4.05, 95% CI 3.28-5.00) in subjects with occult cancer compared with those without cancer. Among subjects with occult cancer, those with VTE had a higher proportion of prothrombotic and advanced cancers than those without VTE. The VTE risk increased according to individual prothrombotic genotypes and GRS in cancer-free subjects, while no such effect was observed in subjects with occult cancer (HR for ≥4 versus ≤1 risk alleles in GRS: 1.14, 95% CI 0.61-2.11). CONCLUSIONS Five well-established prothrombotic genotypes, individually or combined, were not associated with increased risk of VTE in individuals with occult cancer.
Collapse
Affiliation(s)
- Hanne Skille
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Benedikte Paulsen
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway; HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Marianne T Severinsen
- Department of Clinical Medicine, Aalborg University, Denmark; Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway; HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Søren R Kristensen
- Department of Clinical Medicine, Aalborg University, Denmark; Department of Clinical Biochemistry, Aalborg University hospital, Aalborg, Denmark
| | - Inger Anne Næss
- Department of Rheumatology, Trondheim University Hospital, Trondheim, Norway
| | - Kristian Hindberg
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Anne Tjønneland
- Diet, Genes and Environment, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Sigrid K Brækkan
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway.
| | - John-Bjarne Hansen
- Thrombosis Research Center (TREC), Department of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
26
|
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
|
27
|
Ilich A, Kumar V, Henderson M, Mallick R, Wells P, Carrier M, Key NS. Biomarkers in cancer patients at risk for venous thromboembolism: data from the AVERT study. Thromb Res 2021; 191 Suppl 1:S31-S36. [PMID: 32736776 DOI: 10.1016/s0049-3848(20)30394-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The mechanisms surrounding cancer-associated venous thromboembolism (VTE) are not well characterized. AVERT, a randomized placebo controlled thromboprophylaxis study in ambulatory cancer patients, provides a unique opportunity to gain insights into thrombotic mechanism(s). METHODS All available citrated platelet-free plasma samples collected at the point of randomization from individuals enrolled in the AVERT study were evaluated for the expression of D-dimer, soluble P-selectin (sP- selectin), active plasminogen activator inhibitor 1 (aPAI-1), clot lysis time (CLT) and activated factor XIa-C1 inhibitor complex (FXIa-C1). We compared the differential expression of sP-selectin, aPAI-1, CLT and FXIa-C1 among individual tumor types with normal controls. We evaluated the impact of disease type (hematologic versus solid organ malignancy) and stage (metastatic versus non-metastatic) on individual biomarker expression. RESULTS We included 449 AVERT participants in this analysis. Baseline expression of the selected thrombosis biomarkers differed significantly by individual tumor type compared with normal controls. Levels of aPAI-1, CLT, FXIa-C1 and sP-selectin were significantly elevated in individuals with lymphoma compared to individuals with non-metastatic solid organ malignancies (p<0.05). Individuals with metastatic solid organ disease had elevated levels of D-dimer and sP-selectin compared to those with non-metastatic disease (p<0.05). CONCLUSION Among a cohort of ambulatory patients at intermediate to high risk of VTE, these exploratory findings suggest that baseline activation of coagulation and fibrinolysis pathways vary significantly by tumor type and disease stage.
Collapse
Affiliation(s)
- Anton Ilich
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA; These authors contributed equally
| | - Vaibhav Kumar
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA; These authors contributed equally
| | - Michael Henderson
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ranjeeta Mallick
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Philip Wells
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Marc Carrier
- Department of Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Nigel S Key
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
28
|
Wang Q, Cao B, Peng L, Dai W, Jiang Y, Xie T, Fang Q, Wang Y, Wu L, Han Y, Lang J, Mi K. Development and Validation of a Practical Prognostic Coagulation Index for Patients with Esophageal Squamous Cell Cancer. Ann Surg Oncol 2021; 28:8450-8461. [PMID: 34101065 DOI: 10.1245/s10434-021-10239-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND This study aimed to establish an effective and practical prognostic index for esophageal squamous cell cancer (ESCC) based on the coagulation factors. METHODS The training cohort of 965 patients with ESCC was retrospectively collected at Sichuan Cancer Hospital from 2012 to 2014, along with clinical characteristics and follow-up information. Risk factors of coagulation status, including 11 blood parameters (platelet [PLT], mean platelet volume [MPV], platelet distribution width [PDW], plateletocrit [PCT], thrombin time [TT], prothrombin time [PT], international normalized ratio [INR], activated partial thromboplastin time [APTT], fibrinogen, D-dimer, and fibrinogen degradation product [FDP]), were studied by least absolute shrinkage and selection operator (LASSO) Cox regression and the Coagulation Index was established. The index was validated in a cohort of 848 patients with ESCC at the same institution, from 2015 to 2016. RESULTS Three variables of PLT, MPV, and fibrinogen were identified by selecting features with coefficients in the LASSO algorithm, and a Coagulation Index was established as follows: Coagulation Index = 0.0005 × PLT (109/L) - 0.0384 × MPV (fL) + 0.1148 × fibrinogen (g/L). A higher Coagulation Index score was significantly associated with higher pT stage and pN stage (p < 0.05). With this prognostic index, patients could be stratified into three risk groups. The 3-year overall survival (OS) rates of the low-, middle- and high-risk groups in the training cohort were 63.5%, 55.5% and 43.1%, respectively (log-rank p < 0.001). Similarly, in the validation set, the respective 3-year OS for each risk group was significantly different across the three risk groups. Multivariate analysis indicated that the Coagulation Index remained a significant factor for predicting OS, independently of pathological TNM stage. CONCLUSIONS The Coagulation Index is an independent predictor of survival for patients with ESCC.
Collapse
Affiliation(s)
- Qifeng Wang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Radiation Oncology, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bangrong Cao
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Peng
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Dai
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yinchun Jiang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tianpeng Xie
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiang Fang
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yongtao Han
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Radiation Oncology, Sichuan Cancer Hospital and Institution, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kun Mi
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
29
|
Roopkumar J, Swaidani S, Kim AS, Thapa B, Gervaso L, Hobbs BP, Wei W, Alban TJ, Funchain P, Kundu S, Sangwan N, Rayman P, Pavicic PG, Diaz-Montero CM, Barnard J, McCrae KR, Khorana AA. Increased Incidence of Venous Thromboembolism with Cancer Immunotherapy. MED 2021; 2:423-434. [PMID: 34036293 PMCID: PMC8143033 DOI: 10.1016/j.medj.2021.02.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cancer immunotherapy is associated with several immune-related adverse events, but the relationship between immunotherapy and venous thromboembolism has not been thoroughly studied. METHODS We conducted a retrospective cohort study of 1,686 patients who received immunotherapy for a variety of malignancies to determine the incidence of venous thromboembolism and the impact of venous thromboembolism on survival. To examine the potential role of inflammation in venous thromboembolism, we also profiled immune cells and plasma cytokines in blood samples obtained prior to initiation of immunotherapy in a sub-cohort of patients treated on clinical trials who subsequently did (N = 15), or did not (N = 10) develop venous thromboembolism. FINDINGS Venous thromboembolism occurred while on immunotherapy in 404/1686 patients (24%) and was associated with decreased overall survival [HR=1.22 (95% CI 1.06-1.41), p<0.008]. Patients that developed venous thromboembolism had significantly higher pretreatment levels of myeloid-derived suppressor cells (5.382 ± 0.873 vs. 3.341 ± 0.3402, mean ± SEM; p=0.0045), interleukin 8 (221.2 ± 37.53 vs. 111.6 ± 25.36, mean ± SEM; p=0.016), and soluble vascular cell adhesion protein 1 (1210 ± 120.6 vs. 895.5 ± 53.34, mean ± SEM; p=0.0385). CONCLUSIONS These findings demonstrate that venous thromboembolism is an underappreciated and important immune-related adverse event associated with cancer immunotherapy, and may implicate an interleukin 8 and myeloid-derived suppressor cell-driven pathway in pathogenesis.
Collapse
Affiliation(s)
- Joanna Roopkumar
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Shadi Swaidani
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Ann S. Kim
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Bicky Thapa
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Lorenzo Gervaso
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Brian P. Hobbs
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Wei Wei
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Tyler J Alban
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Pauline Funchain
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Suman Kundu
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Naseer Sangwan
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Patricia Rayman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Paul G. Pavicic
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - C. Marcela Diaz-Montero
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Keith R. McCrae
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Alok A. Khorana
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
30
|
Thromboembolism in Malignant Musculoskeletal Tumour: A Literature Review. Adv Orthop 2021; 2021:6678167. [PMID: 33688439 PMCID: PMC7910073 DOI: 10.1155/2021/6678167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/22/2021] [Accepted: 02/09/2021] [Indexed: 12/03/2022] Open
Abstract
Malignant musculoskeletal tumour may cause considerable burden to general health. The fast growth combined with the tumour characteristics and its invasion capability resulted in the poor prognosis of malignant musculoskeletal tumour. Malignant musculoskeletal tumour may cause significant disability by destroying normal tissue that plays important role in body kinematics. Thromboembolism, including deep vein thrombosis, pulmonary embolism, and other kinds of venous thromboembolism, is one of the most underestimated complications of musculoskeletal tumour. Normally, thrombosis ensues when pathologic factors overcame the body hemostatic regulatory capabilities, which will predispose the body to the formation of thrombus. Venous thromboembolism in musculoskeletal tumour may develop as a result of interaction between the tumour pathologic capabilities and its interaction with normal bodily functions. In this study, we reviewed the burden of musculoskeletal tumour and its complication on global health. Then, the review will focus on the pathologic and clinical aspect of thromboembolism in malignant musculoskeletal tumour, including pathophysiology, diagnosis, and treatment based on recent findings and literature.
Collapse
|
31
|
Schaefer JK, Li M, Wu Z, Basu T, Barnes GD, Carrier M, Griggs JJ, Sood SL. Clinical and sociodemographic factors associated with anticoagulant use for cancer associated venous thromboembolism. J Thromb Thrombolysis 2021; 52:214-223. [PMID: 33544284 DOI: 10.1007/s11239-021-02392-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 10/22/2022]
Abstract
Cancer associated thrombosis (CAT) is a leading cause of death among patients with cancer. It is not clear if non-clinical factors are associated with anticoagulation receipt. We conducted a retrospective cohort study of Optum's de-identified Clinformatics® Database of adults with cancer diagnosed between 2009 and 2016 who developed CAT, treated with an outpatient anticoagulant (warfarin, low molecular weight heparin (LMWH), or a direct oral anticoagulant (DOAC)). Of 12,622 patients, three months after an episode of CAT, 1,485 (12%) were on LMWH, 1,546 (12%) on DOACs, and 9,591 (76%) were on warfarin. When controlling for other factors, anticoagulant use was significantly associated with socioeconomic factors, region, co-morbidities, type of thrombosis, and cancer subtype. Patients with a bachelor's degree or greater level of education were less likely to receive warfarin (OR: 0.77; 95% CI: [0.59, 0.99]; p < 0.046) or DOACs (OR: 0.67; 95% CI: [0.55, 0.82]; p < 0.001) compared to LMWH. Patients with higher income levels were more likely to receive LMWH or DOACs compared to warfarin, while patients across all income levels were equally likely to receive LMWH or DOACs. Non-clinical factors including income, education, and region, are associated with anticoagulation receipt three months after an episode of CAT. Sociodemographic factors may result in some patients receiving suboptimal care and contribute to non-guideline concordant care for CAT.
Collapse
Affiliation(s)
- Jordan K Schaefer
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, C366 Med Inn Building, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA.
| | - Mengbing Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Zhenke Wu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, USA.,Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Tanima Basu
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey D Barnes
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA.,Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marc Carrier
- Department of Medicine, The Ottawa Hospital Research Institute at the University of Ottawa, Ottawa, ON, Canada
| | - Jennifer J Griggs
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, C366 Med Inn Building, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA.,Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
| | - Suman L Sood
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, C366 Med Inn Building, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, USA
| |
Collapse
|
32
|
Schaefer JK, Li M, Wu Z, Basu T, Dorsch MP, Barnes GD, Carrier M, Griggs JJ, Sood SL. Anticoagulant medication adherence for cancer-associated thrombosis: A comparison of LMWH to DOACs. J Thromb Haemost 2021; 19:212-220. [PMID: 33104289 DOI: 10.1111/jth.15153] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/20/2022]
Abstract
Essentials It is not clear if patients are less adherent to low molecular weight heparin (LMWH) compared to direct oral anticoagulants (DOACs) for cancer-associated thrombosis (CAT). We evaluated medication adherence among two propensity-matched groups of patients with CAT by comparing the proportion of days covered (PDC). Median treatment persistence on DOACs was more than 80 days longer than LMWH. Medication adherence was high (~95%) and was similar with LMWH compared to DOACs. ABSTRACT: Background Low molecular weight heparin (LMWH) and direct oral anticoagulants (DOACs) are used to treat cancer-associated thrombosis (CAT). It is not clear if patients are less adherent to LMWH compared to DOACs. Objectives To compare medication persistence and adherence between LMWH and DOACs. Patients/Methods We analyzed Optum's de-identified Clinformatics® Data Mart Database of privately insured adults with cancer diagnosed between January 2009 and October 2015 who were undergoing chemotherapy, immunotherapy, targeted or hormonal therapies; developed CAT; and were treated with an outpatient anticoagulant. The proportion of days covered (PDC) was calculated from the date of anticoagulant prescription until the anticoagulant was switched, stopped, or the study end. Medication adherence was defined as PDC ≥ 80%, ≥95%, and by comparing the mean PDC. Results Two propensity-matched groups of 1128 patients were identified. Patient persistence was higher with DOACs compared to LMWH (median 116 days versus 34 days). With adherence defined as PDC ≥ 80%, we found no significant difference (95.6% versus 94.6% adherence with DOACs versus LMWH, P = .33). The mean difference of PDC between the two groups was also similar. With medication adherence defined as PDC ≥ 95%, adherence was evident in 73% of DOAC users and 81% of patients on LMWH (P < .001). Prescription copayments were higher on average for LMWH compared to DOACs (mean $153.61 versus 40.67; standard deviation $306.74 versus $33.11). Conclusion Patients remain on DOACs longer than LMWH, but medication adherence is similar with LMWH.
Collapse
Affiliation(s)
- Jordan K Schaefer
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Mengbing Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Zhenke Wu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, USA
- Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Tanima Basu
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
| | - Michael P Dorsch
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey D Barnes
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Marc Carrier
- Department of Medicine, The Ottawa Hospital Research Institute at the University of Ottawa, ON, Canada
| | - Jennifer J Griggs
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI, USA
| | - Suman L Sood
- Department of Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Chen C, Li J, Li J, Wang X, Wang X, Du N, Ren L. Application of an elevated plasma D-dimer cut-off value improves prognosis prediction of advanced non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1153. [PMID: 33241002 PMCID: PMC7576026 DOI: 10.21037/atm-20-5947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background Tumor-related coagulation dysfunction has been reported to be closely associated with poor prognosis. The present study is aimed to evaluate the prognostic prediction of an elevated plasma D-dimer cut-off value in advanced non-small cell lung cancer (NSCLC). Methods A total of 233 patients initially diagnosed with advanced NSCLC were retrospectively analyzed, an elevated plasma cut-off value 981 ng/mL of D-dimer, which was instead of the clinical cut-off value 500 mg/mL, was used to determine the high and low. Univariate analysis using the Kaplan-Meier method and log-ranking test, and the multivariate analysis using the Cox proportional hazard regression model were performed. Results Results showed when using the D-dimer value of 500 ng/mL as an evaluation standard, there was no significant difference in gender, age, smoking status, histopathology and overall survival rate between normal D-dimer (≤500 ng/mL) and high D-dimer (>500 ng/mL) group. However, when the evaluation standard for plasma D-dimer was set at 981 ng/mL, the age distribution of the high D-dimer (>981 ng/mL) group was significantly different from the normal D-dimer (≤981 ng/mL) group. Moreover, the overall survival rate in the high D-dimer (>981 ng/mL) group was significantly lower than that in the normal D-dimer (≤981 ng/mL) group. Conclusions The present study implied that increasing the plasma D-dimer cut-off value to 981 ng/mL is more beneficial to prognosis prediction in advanced NSCLC.
Collapse
Affiliation(s)
- Chong Chen
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Jianhua Li
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Jing Li
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Xu Wang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Xiaoyan Wang
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Na Du
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Li Ren
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| |
Collapse
|
34
|
Trousseau's syndrome associated with rapidly emerging pancreatic adenocarcinoma soon after esophagectomy: A case report. Int J Surg Case Rep 2020; 77:605-609. [PMID: 33395856 PMCID: PMC7708758 DOI: 10.1016/j.ijscr.2020.11.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 11/20/2022] Open
Abstract
The first reported case of Trousseau’s syndrome associated with rapidly emerging pancreatic cancer potentially triggered esophagectomy. The aggressively emerging pancreatic cancer with mucin production may be a potential mechanism for cancer-related thrombosis. When a patient with cancer encountered small, multiple cerebral infarctions postoperatively, the body should be checked for occult malignancy.
Introduction Trousseau’s syndrome is characterized as an unexpected, cancer-associated thrombotic event. We describe the first reported case of Trousseau’s syndrome associated with rapidly emerging pancreatic cancer potentially triggered by esophagectomy. Presentation of case A 79-year-old asymptomatic male with clinical stage I esophageal squamous cell carcinoma underwent thoracoscopic subtotal esophagectomy. On postoperative day 46, the patient presented with weakness of his left upper extremity due to multiple cerebral and cerebellar infarctions, with no evidence of atherosclerotic or cardiogenic thrombi. An abdominal computed tomography (CT) showed a pancreatic tumor with multiple liver metastases. Extremely high D-dimer and the CT findings suggested Trousseau’s syndrome associated with a rapidly emerging neoplasm as the etiology of the brain infarction. Although further thrombotic events did not occur, his condition deteriorated rapidly and died on the 31st days of onset. The autopsy revealed multiple small infarctions, with multiple thrombi in the cerebral hemispheres, brain stem, and cerebellum. Histological evaluation revealed pancreatic adenocarcinoma with nodal and liver metastases. Discussion A hypercoagulable state associated with the aggressively emerging pancreatic adenocarcinoma, accompanied by cancer cell production of mucin, may be a potential mechanism for cancer-related thrombosis. Conclusion In patients who received intensive surgical treatment and encountered unexplained brain infarctions in the multi-arterial territory, Trousseau’s syndrome should be considered, and investigation for occult malignancy is required.
Collapse
|
35
|
Wilson NR, Khan M, Cox TM, Nassif M, Qiao W, Garg N, Aung FM, Oo TH, Rojas‐Hernandez CM. Bleeding outcomes in thrombocytopenic acute leukemic patients with venous thromboembolism. EJHAEM 2020; 1:448-456. [PMID: 35845011 PMCID: PMC9175819 DOI: 10.1002/jha2.90] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022]
Abstract
Cancer-associated thrombosis in acute leukemia patients with severe thrombocytopenia (platelets ≤50 × 109/L) poses a management challenge due to competing risks of bleeding and recurrent thrombosis. A retrospective analysis was conducted to determine the occurrence of clinically relevant bleeding (CRB) rates during treatment for acute venous thromboembolic events (VTE) in thrombocytopenic acute leukemic patients. A cohort of 74 patients were subgrouped into three VTE-treatment interventions: anticoagulation (n = 24), inferior vena cava filter placement (n = 22), and observation (n = 28). Multivariate analysis found a significant correlation between CRB occurrence and quantity of overall blood transfusions, chemotherapy administration, and relapsed leukemia presentation. There was no difference in the occurrence of CRB between VTE-treatment subgroups, regardless of initial platelet count at the time of VTE diagnosis. Regarding the hematologic parameters, only the velocity of the platelet count recovery was associated with the risk of bleeding. From this analysis, it appears the trajectory of the platelet count and the factors associated with a slower recovery of it, are the main determinants for the occurrence of hemorrhagic complications during VTE treatment in acute leukemia.
Collapse
Affiliation(s)
- Nathaniel R. Wilson
- Department of Internal MedicineThe University of Texas Health Science Center at HoustonHoustonTexas
| | - Maliha Khan
- Department of Hematology and OncologyThe University of Arkansas for Medical SciencesLittle RockArkansas
| | - Travis M. Cox
- Department of Hematology and OncologyThe University of Texas Health Science Center San AntonioSan AntonioTexas
| | - Mohammed Nassif
- Department of Pediatrics – Research Resource OfficeBaylor College of MedicineHoustonTexas
| | - Wei Qiao
- Department of BiostatisticsThe University of Texas M.D. Anderson Cancer CenterHoustonTexas
| | - Naveen Garg
- Department of Diagnostic RadiologyThe University of Texas M.D. Anderson Cancer CenterHoustonTexas
| | - Fleur M. Aung
- Department of Laboratory MedicineThe University of Texas M.D. Anderson Cancer CenterHoustonTexas
| | - Thein Hlaing Oo
- Section of Benign HematologyThe University of Texas M.D. Anderson Cancer CenterHoustonTexas
| | | |
Collapse
|
36
|
Skille H, Paulsen B, Hveem K, Gabrielsen ME, Brumpton B, Hindberg K, Gran OV, Rosendaal FR, Braekkan SK, Hansen JB. Combined effects of five prothrombotic genotypes and cancer on the risk of a first venous thromboembolic event. J Thromb Haemost 2020; 18:2861-2869. [PMID: 32671915 DOI: 10.1111/jth.15011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND The role of combined prothrombotic genotypes in cancer-related venous thromboembolism (VTE) is scarcely studied. We aimed to investigate the impact of a 5-single nucleotide polymorphism (SNP) score on the risk of VTE in patients with and without cancer using a population-based case-cohort. METHODS Cases with a first VTE (n = 1493) and a subcohort (n = 13 072) were derived from the Tromsø Study (1994-2012) and the Nord-Trøndelag Health Study (1995-2008). Five SNPs previously reported as a risk score were genotyped: ABO (rs8176719), F5 (rs6025), F2 (rs1799963), FGG (rs2066865), and F11 (rs2036914). Hazard ratios (HRs) for VTE were estimated according to cancer status and the number of risk alleles in the 5-SNP score (0-1, 2-3, and ≥4 alleles). RESULTS During a median follow-up of 12.3 years, 1496 individuals were diagnosed with cancer, of whom 232 experienced VTE. The VTE risk increased with the number of risk alleles in the 5-SNP score among subjects without and with cancer. In cancer-free subjects, the HR was 2.17 (95% confidence interval [CI] 1.79-2.62) for ≥4 versus 0-1 risk alleles. In cancer patients, the corresponding HR was 1.93 (95% CI 1.28-2.91). The combination of cancer and ≥4 risk alleles yielded a 17-fold (HR 17.1, 95% CI 12.5-23.4) higher risk of VTE compared with cancer-free subjects with 0-1 risk alleles. CONCLUSION The risk of VTE increases with the number of prothrombotic risk alleles in subjects with and without cancer, and the combination of prothrombotic risk alleles and cancer leads to a highly elevated risk of VTE.
Collapse
Affiliation(s)
- Hanne Skille
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Benedikte Paulsen
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Ben Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Kristian Hindberg
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Olga V Gran
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Frits R Rosendaal
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Sigrid K Braekkan
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - John-Bjarne Hansen
- K.G. Jebsen Thrombosis Research and Expertise Center (TREC), Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
37
|
Marchetti M, Giaccherini C, Masci G, Verzeroli C, Russo L, Celio L, Sarmiento R, Gamba S, Tartari CJ, Diani E, Vignoli A, Malighetti P, Spinelli D, Kuderer NM, Nichetti F, Minelli M, Tondini C, Barni S, Giuliani F, Petrelli F, D'Alessio A, Gasparini G, Labianca R, Santoro A, De Braud F, Falanga A. Thrombin generation predicts early recurrence in breast cancer patients. J Thromb Haemost 2020; 18:2220-2231. [PMID: 32397009 DOI: 10.1111/jth.14891] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cancer patients present with a hypercoagulable state often associated with poor disease prognosis. OBJECTIVES This study aims to evaluate whether thrombin generation (TG), a global coagulation test, may be a useful tool to improve the identification of patients at high risk of early disease recurrence (ie, E-DR within 2 years) after breast cancer surgery. PATIENTS/METHODS A cohort of 522 newly diagnosed patients with surgically resected high-risk breast cancer were enrolled in the ongoing prospective HYPERCAN study. TG potential was measured in plasma samples collected before starting systemic chemotherapy. Significant predictive hemostatic and clinic-pathological parameters were identified in the derivation cohort by Cox regression analysis. A risk prognostic score for E-DR was generated in the derivation and tested in the validation cohort. RESULTS After a median observation period of 3.4 years, DR occurred in 51 patients, 28 of whom were E-DR. E-DR subjects presented with the highest TG values as compared to both late-DR (from 2 to 5 years) and no relapse subjects (P < .01). Multivariate analysis in the derivation cohort identified TG, mastectomy, triple negative and Luminal B HER2-neg molecular subtypes as significant independent predictors for E-DR, which were utilized to generate a risk assessment score. In the derivation and validation cohorts, E-DR rates were 2.3% and 0% in the low-risk, 10.1% and 6.3% in the intermediate-risk, and 18.2% and 16.7%, in the high-risk categories, respectively. CONCLUSIONS Inclusion of TG in a risk-assessment model for E-DR significantly helps the identification of operated breast cancer patients at high risk of very early relapse.
Collapse
Affiliation(s)
- Marina Marchetti
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Cinzia Giaccherini
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Giovanna Masci
- Oncology Unit, IRCCS Humanitas Institute, Rozzano, Italy
| | - Cristina Verzeroli
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Laura Russo
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Luigi Celio
- Oncology Unit, IRCCS National Cancer Institute, Milan, Italy
| | | | - Sara Gamba
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Carmen J Tartari
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Erika Diani
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Alfonso Vignoli
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Malighetti
- Department of Management, Information and Production Engineering, University of Bergamo, Bergamo, Italy
| | - Daniele Spinelli
- Department of Management, Information and Production Engineering, University of Bergamo, Bergamo, Italy
| | | | | | - Mauro Minelli
- Oncology Unit, Hospital San Giovanni Addolorata, Rome, Italy
| | - Carlo Tondini
- Oncology Unit, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Sandro Barni
- Oncology Unit, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | | | - Fausto Petrelli
- Oncology Unit, Hospital Treviglio-Caravaggio, Treviglio, Italy
| | - Andrea D'Alessio
- Medical Oncology and Internal Medicine, Policlinico San Marco, Bergamo, Italy
| | | | - Roberto Labianca
- Department Oncology Bergamo Province, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Anna Falanga
- Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
- School of Medicine, University of Milan Bicocca, Italy
| |
Collapse
|
38
|
Meikle CK, Meisler AJ, Bird CM, Jeffries JA, Azeem N, Garg P, Crawford EL, Kelly CA, Gao TZ, Wuescher LM, Willey JC, Worth RG. Platelet-T cell aggregates in lung cancer patients: Implications for thrombosis. PLoS One 2020; 15:e0236966. [PMID: 32776968 PMCID: PMC7416940 DOI: 10.1371/journal.pone.0236966] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
Platelet-leukocyte aggregates (PLAs) are associated with increased thrombosis risk. The influence of PLA formation is especially important for cancer patients, since thrombosis accounts for approximately 10% of cancer-associated deaths. Our objective was to characterize and quantify PLAs in whole blood samples from lung cancer patients compared to healthy volunteers with the intent to analyze PLA formation in the context of lung cancer-associated thrombosis. Consenting lung cancer patients (57) and healthy volunteers (56) were enrolled at the Dana Cancer Center at the University of Toledo Health Science Campus. Peripheral blood samples were analyzed by flow cytometry. Patient medical history was reviewed through electronic medical records. Most importantly, we found lung cancer patients to have higher percentages of platelet-T cell aggregates (PTCAs) than healthy volunteers among both CD4+ T lymphocyte and CD8+ T lymphocyte populations. Our findings demonstrate that characterization of PTCAs may have clinical utility in differentiating lung cancer patients from healthy volunteers and stratifying lung cancer patients by history of thrombosis.
Collapse
Affiliation(s)
- Claire K. Meikle
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Adam J. Meisler
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Cara M. Bird
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Joseph A. Jeffries
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Nabila Azeem
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Priyanka Garg
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Erin L. Crawford
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Clare A. Kelly
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Tess Z. Gao
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Leah M. Wuescher
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - James C. Willey
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| | - Randall G. Worth
- Department of Medical Microbiology & Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States of America
| |
Collapse
|
39
|
Pilli VS, Datta A, Dorsey A, Liu B, Majumder R. Modulation of protein S and growth arrest specific 6 protein signaling inhibits pancreatic cancer cell survival and proliferation. Oncol Rep 2020; 44:1322-1332. [PMID: 32945517 PMCID: PMC7448444 DOI: 10.3892/or.2020.7689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Thrombotic complications and hypercoagulopathies are commonly associated with the progression of pancreatic ductal adenocarcinoma (PDAC). Although the mechanistic link between the two phenomena is uncertain, there is evidently an increase in procoagulant proteins and a decrease in anticoagulants in PDAC patients. For example, the anticoagulant protein S (PS) is decreased during the progression of PDAC, a condition that possibly contributes to the hypercoagulopathies. PS is also an important signaling molecule that binds a family of tyrosine kinase receptors known as TAM (Tyro3, Axl and Mer) receptors; TAM receptors are often upregulated in different cancers. Growth Arrest Specific 6 or GAS6 protein, a homolog of PS, is also a TAM receptor family ligand. The downstream signaling pathways triggered by this ligand-receptor interaction perform diverse functions, such as cell survival, proliferation, efferocytosis, and apoptosis. Targeting the TAM receptors to treat cancer has had limited success; side effects are a significant obstacle due to the widespread numerous functions of TAM receptors. In the present study, it was revealed that PS-TAM interaction was pro-apoptotic, whereas GAS6-mediated TAM signaling promoted proliferation and survival in select PDAC cell lines. Furthermore, by regulating the balance between these two signaling pathways (by overexpressing PS or knocking down GAS6), the proliferative potential of the cells was decreased. Both long-term and short-term effects of natural PS overexpression were comparable to the treatment of the cells with the drug UNC2025, which inhibits the Mer-receptor. The present study lays the foundation for investigation of PS as a therapeutic agent to control cancer progression and to concurrently arrest thrombotic events.
Collapse
Affiliation(s)
- Vijaya S Pilli
- Department of Biochemistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Arani Datta
- Department of Biochemistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Adrianne Dorsey
- Department of Biochemistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Bo Liu
- Department of Surgery, University of Wisconsin, Madison, WI 53705, USA
| | - Rinku Majumder
- Department of Biochemistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
40
|
Welsh JD, Hoofnagle MH, Bamezai S, Oxendine M, Lim L, Hall JD, Yang J, Schultz S, Engel JD, Kume T, Oliver G, Jimenez JM, Kahn ML. Hemodynamic regulation of perivalvular endothelial gene expression prevents deep venous thrombosis. J Clin Invest 2020; 129:5489-5500. [PMID: 31710307 DOI: 10.1172/jci124791] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Deep venous thrombosis (DVT) and secondary pulmonary embolism cause approximately 100,000 deaths per year in the United States. Physical immobility is the most significant risk factor for DVT, but a molecular and cellular basis for this link has not been defined. We found that the endothelial cells surrounding the venous valve, where DVTs originate, express high levels of FOXC2 and PROX1, transcription factors known to be activated by oscillatory shear stress. The perivalvular venous endothelial cells exhibited a powerful antithrombotic phenotype characterized by low levels of the prothrombotic proteins vWF, P-selectin, and ICAM1 and high levels of the antithrombotic proteins thrombomodulin (THBD), endothelial protein C receptor (EPCR), and tissue factor pathway inhibitor (TFPI). The perivalvular antithrombotic phenotype was lost following genetic deletion of FOXC2 or femoral artery ligation to reduce venous flow in mice, and at the site of origin of human DVT associated with fatal pulmonary embolism. Oscillatory blood flow was detected at perivalvular sites in human veins following muscular activity, but not in the immobile state or after activation of an intermittent compression device designed to prevent DVT. These findings support a mechanism of DVT pathogenesis in which loss of muscular activity results in loss of oscillatory shear-dependent transcriptional and antithrombotic phenotypes in perivalvular venous endothelial cells, and suggest that prevention of DVT and pulmonary embolism may be improved by mechanical devices specifically designed to restore perivalvular oscillatory flow.
Collapse
Affiliation(s)
- John D Welsh
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark H Hoofnagle
- Department of Surgery, Division of Traumatology, Surgical Critical Care, and Emergency Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sharika Bamezai
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael Oxendine
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Lillian Lim
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joshua D Hall
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Jisheng Yang
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan Schultz
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Tsutomu Kume
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Juan M Jimenez
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Nimblette C, Seecheran R, Kawall J, Seecheran V, Persad S, Ramsaroop K, Seecheran NA. Dissolution of metastatic thymic carcinoma-associated right atrial thrombus with rivaroxaban. SAGE Open Med Case Rep 2020; 8:2050313X20927596. [PMID: 32551115 PMCID: PMC7278298 DOI: 10.1177/2050313x20927596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 04/27/2020] [Indexed: 11/15/2022] Open
Abstract
Thymic carcinoma typically exhibits more clinically aggressive behavior and portends a worse prognosis as compared to thymoma. Venous thromboembolism is a significant cause of morbidity and mortality in oncologic patients. Traditionally, the standard-of-care management of cancer-associated venous thromboembolism has been therapeutic anticoagulation with low molecular weight heparins; however, with the advent of direct oral anticoagulants, there is an ongoing paradigm shift to transition to these novel agents in an attempt to attenuate cancer-associated venous thromboembolism events. We describe an exceedingly rare case of metastatic thymic carcinoma-associated right atrial thrombus with high-risk embolic features, which subsequently underwent near-complete dissolution with rivaroxaban after 3 months.
Collapse
Affiliation(s)
- Curlene Nimblette
- North Central Regional Health Authority, Mt. Hope, Trinidad and Tobago
| | - Rajeev Seecheran
- North Central Regional Health Authority, Mt. Hope, Trinidad and Tobago
| | - Jessica Kawall
- North Central Regional Health Authority, Mt. Hope, Trinidad and Tobago
| | - Valmiki Seecheran
- North Central Regional Health Authority, Mt. Hope, Trinidad and Tobago
| | - Sangeeta Persad
- North Central Regional Health Authority, Mt. Hope, Trinidad and Tobago
| | | | - Naveen Anand Seecheran
- Department of Clinical Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
42
|
Kasthuri RS, Hisada Y, Ilich A, Key NS, Mackman N. Effect of chemotherapy and longitudinal analysis of circulating extracellular vesicle tissue factor activity in patients with pancreatic and colorectal cancer. Res Pract Thromb Haemost 2020; 4:636-643. [PMID: 32548563 PMCID: PMC7292676 DOI: 10.1002/rth2.12317] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION We conducted a longitudinal study in patients with pancreatic and colorectal cancer. We determined the effect of chemotherapy on extracellular vesicle tissue factor (EVTF) activity and the association of plasma EVTF activity with venous thromboembolism (VTE) and survival. MATERIAL AND METHODS We enrolled 13 patients with pancreatic and 22 patients with colorectal cancer. Plasma samples were collected during the 85-day study period. Patients were followed for 3 months after the study period. We recorded symptomatic VTE during the study period (3 months) or asymptomatic deep vein thrombosis detected by ultrasound at day 85. We measured EVTF activity before and after chemotherapy. RESULTS AND CONCLUSIONS In the pancreatic cancer group, 2 patients had elevated levels of EVTF activity. One of these patients developed symptomatic VTE and died, and the second patient did not have a VTE but died. Chemotherapy decreased EVTF activity in 2 pancreatic patients with high levels. In the colorectal cancer group, 4 patients developed VTE, but EVTF activity was not elevated in any patient and no patient died. We observed a borderline significant correlation between EVTF activity and D-dimer in the patients with pancreatic but not colorectal cancer. In this small descriptive study, 2 patients with pancreatic cancer had an elevated level of EVTF activity. Both patients died during the study period, and one had a VTE. Chemotherapy decreased EVTF activity in these patients. In contrast, elevated levels of EVTF activity were not observed in patients with colorectal cancer with or without VTE.
Collapse
Affiliation(s)
- Raj S. Kasthuri
- Division of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Yohei Hisada
- Division of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Anton Ilich
- Division of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Nigel S. Key
- Division of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Nigel Mackman
- Division of Hematology/OncologyDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Blood Research CenterUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| |
Collapse
|
43
|
Nosaka M, Ishida Y, Kimura A, Kuninaka Y, Taruya A, Ozaki M, Tanaka A, Mukaida N, Kondo T. Crucial Involvement of IL-6 in Thrombus Resolution in Mice via Macrophage Recruitment and the Induction of Proteolytic Enzymes. Front Immunol 2020; 10:3150. [PMID: 32117207 PMCID: PMC7019028 DOI: 10.3389/fimmu.2019.03150] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/30/2019] [Indexed: 01/08/2023] Open
Abstract
After the ligation of the inferior vena cava (IVC) of wild-type (WT) mice, venous thrombi formed and grew progressively until 5 days and resolved thereafter. Concomitantly, intrathrombotic gene expression of Il6 was enhanced later than 5 days after IVC ligation. IL-6 protein expression was detected mainly in F4/80-positive macrophages in thrombus. When Il6-deficient (Il6-/-) mice were treated in the same manner, thrombus mass was significantly larger than in WT mice. Moreover, the recovery of thrombosed IVC blood flow was markedly delayed in Il6-/- compared with WT mice. F4/80-positive macrophages in thrombus expressed proteolytic enzymes such as matrix metalloproteinase (Mmp) 2, Mmp9, and urokinase-type plasminogen activator (Plau); and their mRNA expression was significantly reduced in Il6-/- mice. Consistently, the administration of anti-IL-6 antibody delayed the thrombus resolution in WT mice, whereas IL-6 administration accelerated thrombus resolution in WT and Il6-/- mice. Moreover, IL-6 in vitro enhanced Mmp2, Mmp9, and Plau mRNA expression in WT-derived peritoneal macrophages in a dose-dependent manner; and the enhancement was abrogated by a specific Stat3 inhibitor, Stattic. Thus, IL-6/Stat3 signaling pathway can promote thrombus resolution by enhancing Mmp2, Mmp9, and Plau expression in macrophages.
Collapse
Affiliation(s)
- Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akira Taruya
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mitsunori Ozaki
- Department of Neurological Surgery, Wakayama Medical University, Wakayama, Japan
| | - Atushi Tanaka
- Department of Cardiovascular Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
44
|
Dou F, Zhang Y, Yi J, Zhu M, Zhang S, Zhang D, Zhang Y. Association of ALK rearrangement and risk of venous thromboembolism in patients with non-small cell lung cancer: A prospective cohort study. Thromb Res 2019; 186:36-41. [PMID: 31864154 DOI: 10.1016/j.thromres.2019.12.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/08/2019] [Accepted: 12/14/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Isolated reports are inconsistent regarding the risk of venous thromboembolism (VTE) in patients with anaplastic lymphoma kinase (ALK) rearranged non-small cell lung cancer (NSCLC). This study examined whether ALK rearrangement could have an influence on VTE in a prospective cohort. METHODS In a cohort of 836 consecutive patients with NSCLC, patients with epidermal growth factor receptor (EGFR) or kitten rat sarcoma (KRAS) mutations were ruled out for VTE interference. Finally, 341 qualified patients were observed. The median follow up period is 7.5 months (3.1-15.4m). ALK rearrangement was detected by fluorescence in situ hybridization at baseline. RESULTS Overall VTE events occurred in 37 (10.9%) of 341 patients. In multivariable analysis including age, sex, tumor histology, tumor stage, performance status, and ALK status, ALK rearrangement (sub-distribution hazard radio 2.47, 95% confidence interval 1.04-5.90) was associated with the increased risk of VTE. The cumulative incidence of VTE was 26.9% and 9.2% in the patients with and without ALK rearrangement after 6 months. After 1 year the corresponding cumulative incidence was 26.9% and 9.7% respectively (Gray test P = .005). CONCLUSIONS The presence of ALK rearrangement is associated with increased risk of VTE in patients with NSCLC.
Collapse
Affiliation(s)
- Feifei Dou
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Yuan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Jiawen Yi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Di Zhang
- Department of Clinical Epidemiology, 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 Institute of Respiratory Medicine, Beijing 100020, China.
| |
Collapse
|
45
|
Heenkenda MK, Malmström A, Lysiak M, Mudaisi M, Bratthäll C, Milos P, Strandeus M, Åkesson L, Söderkvist P, Uppugunduri S, Osman A. Assessment of genetic and non-genetic risk factors for venous thromboembolism in glioblastoma - The predictive significance of B blood group. Thromb Res 2019; 183:136-142. [PMID: 31677594 DOI: 10.1016/j.thromres.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/23/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Venous thromboembolism (VTE) is a common problem among patients with glioblastoma multiforme (GBM) and with some other cancers. Here, we evaluated genetic and non-genetic potential risk factors for VTE among GBM patients. MATERIALS AND METHODS A cohort of 139 patients treated with concomitant radiotherapy and temozolomide were included in the study. Next generation sequencing and genotyping approaches were applied to assess genetic risk factors in the haemostatic system. Clinical data including surgery, reoperation as well as blood group and patient information such as age and gender were available from patient records. Logistic regression analysis was performed to asses VTE risk. RESULTS In the study 47 patients (34%) were diagnosed for VTE during the course of their disease. When genetic and non-genetic potential risk factors were evaluated, only B blood group was found to be significantly associated with VTE incidence (odds ratio [OR] = 6.91; confidence interval [CI] = 2.19-24.14; P = 0.001). In contrast, A and O blood groups did not correlate with VTE risk. Frontal lobe tumor location also seemed to slightly increase VTE risk compared to other brain sites (OR = 3.14; CI = 1.1-10.7) although the significance level was at borderline (P = 0.05). Current study identified B blood group as the component in non-O blood groups that is responsible for increased VTE risk. CONCLUSION In conclusion, these results suggest for the first time that B blood group is predictive for VTE incidence among patients with glioblastoma, information that may be potentially valuable when selecting GBM patients who are at risk for VTE for anticoagulant prophylaxis.
Collapse
Affiliation(s)
- Menikae K Heenkenda
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Annika Malmström
- Department of Advanced Home Care and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Malgorzata Lysiak
- Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Munila Mudaisi
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Peter Milos
- Department of Neurosurgery and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Lisa Åkesson
- Regional Cancer Centre South East Sweden and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Peter Söderkvist
- Cell Biology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Srinivas Uppugunduri
- Regional Cancer Centre South East Sweden and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Abdimajid Osman
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
| |
Collapse
|
46
|
Five 11α, 12α-epoxy pentacyclic triterpenoid saponins with antithrombus activities from Glechoma longituba. Fitoterapia 2019; 138:104345. [DOI: 10.1016/j.fitote.2019.104345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/20/2019] [Accepted: 08/25/2019] [Indexed: 12/29/2022]
|
47
|
Gomes T, Várady CBS, Lourenço AL, Mizurini DM, Rondon AMR, Leal AC, Gonçalves BS, Bou-Habib DC, Medei E, Monteiro RQ. IL-1β Blockade Attenuates Thrombosis in a Neutrophil Extracellular Trap-Dependent Breast Cancer Model. Front Immunol 2019; 10:2088. [PMID: 31552036 PMCID: PMC6737452 DOI: 10.3389/fimmu.2019.02088] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer patients are at increased risk of developing thrombosis, comorbidity that has been associated with increased neutrophil counts and the formation of neutrophil extracellular traps (NETs). Interleukin-1β (IL-1β) modulates the expression of granulocyte colony-stimulating factor (G-CSF), a cytokine that promotes cancer-associated neutrophilia and NET generation. Herein, we combined a murine breast cancer model with a flow-restriction thrombosis model to evaluate whether the IL-1β blockade could interfere with cancer-associated thrombosis. Mice bearing metastatic 4T1 tumors exhibited high neutrophil counts as well as elevated expression of G-CSF and IL-1β in their tumors. On the other hand, mice bearing non-metastatic 67NR tumors showed no elevation in neutrophil counts and displayed low expression levels of G-CSF and IL-1β in their tumors. 4T1 tumor-bearing mice but not 67NR tumor-bearing mice exhibited a NET-dependent prothrombotic state. Pharmacological blockade of IL-1 receptor (IL-1R) decreased the primary growth of 4T1 tumors and reduced the systemic levels of myeloperoxidase, cell-free DNA (cfDNA) and G-CSF, without interfering with the neutrophil counts. Most remarkably, the blockade of IL-1R abolished the prothrombotic state observed in 4T1 tumor-bearing mice. Overall, our results demonstrate that IL-1β might be a feasible target to attenuate cancer-associated thrombosis, particularly in cancer types that rely on increased G-CSF production and involvement of NET formation.
Collapse
Affiliation(s)
- Tainá Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina B S Várady
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André L Lourenço
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniella M Mizurini
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Araci M R Rondon
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana C Leal
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Barbara S Gonçalves
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil
| | - Dumith Chequer Bou-Habib
- Laboratory on Thymus Research, Oswaldo Cruz Institute/Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| | - Emiliano Medei
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Q Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
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
|
49
|
Bennett JA, Ture SK, Schmidt RA, Mastrangelo MA, Cameron SJ, Terry LE, Yule DI, Morrell CN, Lowenstein CJ. Acetylcholine Inhibits Platelet Activation. J Pharmacol Exp Ther 2019; 369:182-187. [PMID: 30765424 PMCID: PMC6439456 DOI: 10.1124/jpet.118.253583] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/12/2019] [Indexed: 12/18/2022] Open
Abstract
Platelets are key mediators of thrombosis. Many agonists of platelet activation are known, but fewer endogenous inhibitors of platelets, such as prostacyclin and nitric oxide (NO), have been identified. Acetylcholinesterase inhibitors, such as donepezil, can cause bleeding in patients, but the underlying mechanisms are not well understood. We hypothesized that acetylcholine is an endogenous inhibitor of platelets. We measured the effect of acetylcholine or analogs of acetylcholine on human platelet activation ex vivo. Acetylcholine and analogs of acetylcholine inhibited platelet activation, as measured by P-selectin translocation and glycoprotein IIb IIIa conformational changes. Conversely, we found that antagonists of the acetylcholine receptor, such as pancuronium, enhance platelet activation. Furthermore, drugs inhibiting acetylcholinesterase, such as donepezil, also inhibit platelet activation, suggesting that platelets release acetylcholine. We found that NO mediates acetylcholine inhibition of platelets. Our data suggest that acetylcholine is an endogenous inhibitor of platelet activation. The cholinergic system may be a novel target for antithrombotic therapies.
Collapse
Affiliation(s)
- John A Bennett
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Sara K Ture
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Rachel A Schmidt
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Michael A Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Scott J Cameron
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Lara E Terry
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - David I Yule
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Craig N Morrell
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| | - Charles J Lowenstein
- Aab Cardiovascular Research Institute, Department of Medicine (J.A.B., S.K.T., R.A.S., M.A.M., S.J.C., C.N.M., C.J.L.) and Department of Pharmacology and Physiology (L.E.T., D.I.Y.), University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
50
|
Brenner B, Hull R, Arya R, Beyer-Westendorf J, Douketis J, Elalamy I, Imberti D, Zhai Z. Evaluation of unmet clinical needs in prophylaxis and treatment of venous thromboembolism in high-risk patient groups: cancer and critically ill. Thromb J 2019; 17:6. [PMID: 31011294 PMCID: PMC6466798 DOI: 10.1186/s12959-019-0196-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
Background Clinical practice shows that venous thromboembolism (VTE) presents a substantial burden in medical patients, and awareness and advocacy for its primary and secondary prevention remains inadequate. Specific patient populations, such as those with cancer and the critically ill, show elevated risk for VTE, bleeding or both, and significant gaps in VTE prophylaxis and treatment exist in these groups. Objective To present novel insights and consolidated evidence collected from experts, clinical practice guidelines and original studies on the unmet needs in thromboprophylaxis, and on the treatment of VTE in two high-risk patient groups: patients with cancer and the critically ill. Methodology To identify specific unmet needs in the management of VTE, a methodology was designed and implemented that assessed gaps in prophylaxis and treatment of VTE through interviews with 44 experts in the field of thrombosis and haemostasis, and through a review of current guidelines and seminal studies to substantiate the insights provided by the experts. The research findings were then analysed, discussed and consolidated by a multidisciplinary group of experts. Results The gap analysis methodology identified shortcomings in the VTE risk assessment tools, patient stratification approaches for prophylaxis, and the suboptimal use of anticoagulants for primary prophylaxis and treatment. Conclusions Specifically, patients with cancer need better VTE risk assessment tools to tailor primary thromboprophylaxis to tumour types and disease stages, and the potential for drug–drug interactions needs to be considered. In critically ill patients, unfractionated heparin is not advised as a first-line treatment option, and the strength of evidence is increasing for direct oral anticoagulants as a treatment option over low-molecular-weight heparins. Electronic supplementary material The online version of this article (10.1186/s12959-019-0196-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benjamin Brenner
- 1Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Russell Hull
- 2Foothills Medical Centre and Thrombosis Research Unit, University of Calgary, Calgary, Canada
| | - Roopen Arya
- 3King's Thrombosis Centre, Department of Haematological Medicine, King's College Hospital NHS Foundation Trust, London, UK
| | - Jan Beyer-Westendorf
- 3King's Thrombosis Centre, Department of Haematological Medicine, King's College Hospital NHS Foundation Trust, London, UK.,4Thrombosis Research Unit, Department of Medicine I, Division Hematology, University Hospital 'Carl Gustav Carus' Dresden, Dresden, Germany
| | - James Douketis
- 5Department of Medicine, McMaster University, Hamilton, Ontario Canada.,6Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario Canada
| | - Ismail Elalamy
- 7Hematology and Thrombosis Center, Tenon University Hospital, Sorbonne University, Paris, France
| | - Davide Imberti
- 8Haemostasis and Thrombosis Center, Hospital of Piacenza, Piacenza, Italy
| | - Zhenguo Zhai
- 9Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| |
Collapse
|