1
|
Falanga A, Lorusso D, Colombo N, Cormio G, Cosmi B, Scandurra G, Zanagnolo V, Marietta M. Gynecological Cancer and Venous Thromboembolism: A Narrative Review to Increase Awareness and Improve Risk Assessment and Prevention. Cancers (Basel) 2024; 16:1769. [PMID: 38730721 PMCID: PMC11083004 DOI: 10.3390/cancers16091769] [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: 02/14/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The prevention and appropriate management of venous thromboembolism in cancer patients is of paramount importance. However, the literature data report an underestimation of this major problem in patients with gynecological cancers, with an inconsistent venous thromboembolism risk assessment and prophylaxis in this patient setting. This narrative review provides a comprehensive overview of the available evidence regarding the management of venous thromboembolism in cancer patients, focusing on the specific context of gynecological tumors, exploring the literature discussing risk factors, risk assessment, and pharmacological prophylaxis. We found that the current understanding and management of venous thromboembolism in gynecological malignancy is largely based on studies on solid cancers in general. Hence, further, larger, and well-designed research in this area is needed.
Collapse
Affiliation(s)
- Anna Falanga
- Department of Medicine and Surgery, University of Milan-Bicocca, 20900 Monza, Italy; (A.F.); (N.C.)
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Domenica Lorusso
- Fondazione Policlinico Universitario A. Gemelli, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Nicoletta Colombo
- Department of Medicine and Surgery, University of Milan-Bicocca, 20900 Monza, Italy; (A.F.); (N.C.)
- Gynecologic Oncology Program, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
- Department of Interdisciplinary Medicine (DIM), University “A. Moro”, 70124 Bari, Italy
| | - Benilde Cosmi
- Angiology and Blood Coagulation Unit, Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
- Angiology and Blood Coagulation Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giuseppa Scandurra
- Unità Operativa Oncologia Medica, Ospedale Cannizzaro di Catania, 95126 Catania, Italy;
| | | | - Marco Marietta
- Hematology Unit, Azienda Ospedaliero-Universitaria, 41125 Modena, Italy;
| |
Collapse
|
2
|
Erhart F, Widhalm G, Kiesel B, Hackl M, Diendorfer A, Preusser M, Rössler K, Thaler J, Pabinger I, Ay C, Riedl J. The plasma miRNome and venous thromboembolism in high-grade glioma: miRNA Sequencing of a nested case-control cohort. J Cell Mol Med 2024; 28:e18149. [PMID: 38613361 PMCID: PMC11015389 DOI: 10.1111/jcmm.18149] [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: 08/27/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 04/14/2024] Open
Abstract
Patients with high-grade gliomas are at high risk of venous thromboembolism (VTE). MicroRNAs (miRNAs) are small non-coding RNAs with multiple roles in tumour biology, haemostasis and platelet function. Their association with VTE risk in high-grade glioma has not been comprehensively mapped so far. We thus conducted a nested case-control study within 152 patients with WHO grade IV glioma that had been part of a prospective cohort study on VTE risk factors. At inclusion a single blood draw was taken, and patients were thereafter followed for a maximum of 2 years. During that time, 24 patients (16%) developed VTE. Of the other 128 patients, we randomly selected 24 age- and sex-matched controls. After quality control, the final group size was 21 patients with VTE during follow-up and 23 without VTE. Small RNA next-generation sequencing of plasma was performed. We observed that hsa-miR-451a was globally the most abundant miRNA. Notably, 51% of all miRNAs showed a correlation with platelet count. The analysis of miRNAs differentially regulated in VTE patients-with and without platelet adjustment-identified potential VTE biomarker candidates such as has-miR-221-3p. Therewith, we here provide one of the largest and deepest peripheral blood miRNA datasets of high-grade glioma patients so far, in which we identified first VTE biomarker candidates that can serve as the starting point for future research.
Collapse
Affiliation(s)
- Friedrich Erhart
- Department of NeurosurgeryMedical University of ViennaViennaAustria
| | - Georg Widhalm
- Department of NeurosurgeryMedical University of ViennaViennaAustria
| | - Barbara Kiesel
- Department of NeurosurgeryMedical University of ViennaViennaAustria
| | | | | | - Matthias Preusser
- Clinical Division of OncologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - Karl Rössler
- Department of NeurosurgeryMedical University of ViennaViennaAustria
| | - Johannes Thaler
- Clinical Division of Haematology and HaemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - Ingrid Pabinger
- Clinical Division of Haematology and HaemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - Cihan Ay
- Clinical Division of Haematology and HaemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| | - Julia Riedl
- Clinical Division of Haematology and HaemostaseologyDepartment of Medicine IMedical University of ViennaViennaAustria
| |
Collapse
|
3
|
Dasgupta P, Ou A, Lin H, Gregory T, Alfaro-Munoz KD, Yuan Y, Afshar-Khargan V, Kamiya-Matsuoka C, Rousseau JF, Majd NK. The risk and burden of thromboembolic and hemorrhagic events in patients with malignant gliomas receiving bevacizumab. J Neurooncol 2024:10.1007/s11060-023-04551-9. [PMID: 38372903 DOI: 10.1007/s11060-023-04551-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 12/20/2023] [Indexed: 02/20/2024]
Abstract
PURPOSE Bevacizumab has evolved as an integral treatment option for patients with high-grade gliomas. Little is known about clinical risk factors that predispose patients with high-grade gliomas receiving bevacizumab to VTE or ICH. We sought to characterize the clinical risk factors associated with risk of either event. METHODS In this multi-institutional retrospective study, we first evaluated patients with high-grade gliomas who were treated with bevacizumab at University of Texas MD Anderson Cancer Center from 2015-2021. We compared clinical and treatment-related factors among three cohorts: those who developed VTE, ICH, or neither. We further compared survival outcomes of these patients from the time of bevacizumab initiation. Then to further confirm our results in a non-cancer center hospital setting we evaluated patients from two Ascension Seton Hospitals in Austin, Texas which are affiliated with Dell Medical School at the University of Texas at Austin from 2017-2022. RESULTS We found that the presence of cerebral macrobleeding, defined as a magnetic susceptibility of > 1 cm3 on magnetic resonance imaging, was highly associated with risk of developing ICH after initiation of bevacizumab. Development of ICH was significantly associated with poorer survival outcomes. We did not find a statistically significant effect of VTE on survival after bevacizumab initiation. CONCLUSION In order to stratify the risk for developing ICH before the initiation of bevacizumab, we recommend to assess for the presence of cerebral macrobleeding as it is associated with ICH development.
Collapse
Affiliation(s)
- Pushan Dasgupta
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Ou
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy Gregory
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin D Alfaro-Munoz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vahid Afshar-Khargan
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Justin F Rousseau
- Department of Neurology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Nazanin K Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Tawil N, Mohammadnia A, Rak J. Oncogenes and cancer associated thrombosis: what can we learn from single cell genomics about risks and mechanisms? Front Med (Lausanne) 2023; 10:1252417. [PMID: 38188342 PMCID: PMC10769496 DOI: 10.3389/fmed.2023.1252417] [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: 07/03/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Single cell analysis of cancer cell transcriptome may shed a completely new light on cancer-associated thrombosis (CAT). CAT causes morbid, and sometimes lethal complications in certain human cancers known to be associated with high risk of venous thromboembolism (VTE), pulmonary embolism (PE) or arterial thromboembolism (ATE), all of which worsen patients' prognosis. How active cancers drive these processes has long evaded scrutiny. While "unspecific" microenvironmental effects and consequences of patient care (e.g., chemotherapy) have been implicated in pathogenesis of CAT, it has also been suggested that oncogenic pathways driven by either genetic (mutations), or epigenetic (methylation) events may influence the coagulant phenotype of cancer cells and stroma, and thereby modulate the VTE/PE risk. Consequently, the spectrum of driver events and their downstream effector mechanisms may, to some extent, explain the heterogeneity of CAT manifestations between cancer types, molecular subtypes, and individual cases, with thrombosis-promoting, or -protective mutations. Understanding this molecular causation is important if rationally designed countermeasures were to be deployed to mitigate the clinical impact of CAT in individual cancer patients. In this regard, multi-omic analysis of human cancers, especially at a single cell level, has brought a new meaning to concepts of cellular heterogeneity, plasticity, and multicellular complexity of the tumour microenvironment, with profound and still relatively unexplored implications for the pathogenesis of CAT. Indeed, cancers may contain molecularly distinct cellular subpopulations, or dynamic epigenetic states associated with different profiles of coagulant activity. In this article we discuss some of the relevant lessons from the single cell "omics" and how they could unlock new potential mechanisms through which cancer driving oncogenic lesions may modulate CAT, with possible consequences for patient stratification, care, and outcomes.
Collapse
Affiliation(s)
- Nadim Tawil
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Abdulshakour Mohammadnia
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Rue University, Montreal, QC, Canada
| | - Janusz Rak
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
5
|
Woods RR, Lesser GJ. Management of Thromboembolic Disease in Patients with Primary and Metastatic Brain Tumors. Curr Treat Options Oncol 2023; 24:1293-1303. [PMID: 37407888 PMCID: PMC10477143 DOI: 10.1007/s11864-023-01116-w] [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] [Accepted: 05/24/2023] [Indexed: 07/07/2023]
Abstract
OPINION STATEMENT Patients with primary brain tumors are at a substantially elevated risk of venous thromboembolism (VTE) compared to other disease states or other forms of malignancy. Deep venous thrombosis (DVT) and pulmonary embolism (PE), often complicate the care of patients with primary brain tumors, and treatment may pose specific unique risks and considerations for management. This paper critically reviews the relevant literature and the most common treatment options in addition to a discussion regarding the relative risk considerations for neurooncology patients facing thromboembolic disease.
Collapse
Affiliation(s)
- Ryan R. Woods
- Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA
| | - Glenn J. Lesser
- Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA
| |
Collapse
|
6
|
Jo J, Diaz M, Horbinski C, Mackman N, Bagley S, Broekman M, Rak J, Perry J, Pabinger I, Key NS, Schiff D. Epidemiology, biology, and management of venous thromboembolism in gliomas: An interdisciplinary review. Neuro Oncol 2023; 25:1381-1394. [PMID: 37100086 PMCID: PMC10398809 DOI: 10.1093/neuonc/noad059] [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] [Indexed: 04/28/2023] Open
Abstract
Patients with diffuse glioma are at high risk of developing venous thromboembolism (VTE) over the course of the disease, with up to 30% incidence in patients with glioblastoma (GBM) and a lower but nonnegligible risk in lower-grade gliomas. Recent and ongoing efforts to identify clinical and laboratory biomarkers of patients at increased risk offer promise, but to date, there is no proven role for prophylaxis outside of the perioperative period. Emerging data suggest a higher risk of VTE in patients with isocitrate dehydrogenase (IDH) wild-type glioma and the potential mechanistic role of IDH mutation in the suppression of production of the procoagulants tissue factor and podoplanin. According to published guidelines, therapeutic anticoagulation with low molecular weight heparin (LMWH) or alternatively, direct oral anticoagulants (DOACs) in patients without increased risk of gastrointestinal or genitourinary bleeding is recommended for VTE treatment. Due to the elevated risk of intracranial hemorrhage (ICH) in GBM, anticoagulation treatment remains challenging and at times fraught. There are conflicting data on the risk of ICH with LMWH in patients with glioma; small retrospective studies suggest DOACs may convey lower ICH risk than LMWH. Investigational anticoagulants that prevent thrombosis without impairing hemostasis, such as factor XI inhibitors, may carry a better therapeutic index and are expected to enter clinical trials for cancer-associated thrombosis.
Collapse
Affiliation(s)
- Jasmin Jo
- Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University, Greenville, NC, USA
| | - Maria Diaz
- Department of Neurology, Division of Neuro-Oncology, Columbia University, New York, NY, USA
| | - Craig Horbinski
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Nigel Mackman
- Department of Medicine and UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Stephen Bagley
- Department of Medicine, University of Pennsylvania, Philadelphia PA, USA
| | - Marika Broekman
- Department of Neurosurgery, University Medical Center, Utrecht, The Netherlands
| | - Janusz Rak
- Department of Pediatrics, McGill University, Montreal, Canada
| | - James Perry
- Department of Neurology, Sunnybrook Health Sciences Center, Toronto, Canada
| | - Ingrid Pabinger
- Department of Medicine, Medical University of Vienna, Vienna, Austria
| | - Nigel S Key
- Department of Medicine and UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - David Schiff
- Department of Neurology, Division of Neuro-Oncology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Mantha S, Chatterjee S, Singh R, Cadley J, Poon C, Chatterjee A, Kelly D, Sterpi M, Soff G, Zwicker J, Soria J, Ruiz M, Muñoz A, Arcila M. Application of Machine Learning to the Prediction of Cancer-Associated Venous Thromboembolism. RESEARCH SQUARE 2023:rs.3.rs-2870367. [PMID: 37214902 PMCID: PMC10197737 DOI: 10.21203/rs.3.rs-2870367/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Venous thromboembolism (VTE) is a common and impactful complication of cancer. Several clinical prediction rules have been devised to estimate the risk of a thrombotic event in this patient population, however they are associated with limitations. We aimed to develop a predictive model of cancer-associated VTE using machine learning as a means to better integrate all available data, improve prediction accuracy and allow applicability regardless of timing for systemic therapy administration. A retrospective cohort was used to fit and validate the models, consisting of adult patients who had next generation sequencing performed on their solid tumor for the years 2014 to 2019. A deep learning survival model limited to demographic, cancer-specific, laboratory and pharmacological predictors was selected based on results from training data for 23,800 individuals and was evaluated on an internal validation set including 5,951 individuals, yielding a time-dependent concordance index of 0.72 (95% CI = 0.70-0.74) for the first 6 months of observation. Adapted models also performed well overall compared to the Khorana Score (KS) in two external cohorts of individuals starting systemic therapy; in an external validation set of 1,250 patients, the C-index was 0.71 (95% CI = 0.65-0.77) for the deep learning model vs 0.66 (95% CI = 0.59-0.72) for the KS and in a smaller external cohort of 358 patients the C-index was 0.59 (95% CI = 0.50-0.69) for the deep learning model vs 0.56 (95% CI = 0.48-0.64) for the KS. The proportions of patients accurately reclassified by the deep learning model were 25% and 26% respectively. In this large cohort of patients with a broad range of solid malignancies and at different phases of systemic therapy, the use of deep learning resulted in improved accuracy for VTE incidence predictions. Additional studies are needed to further assess the validity of this model.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gerald Soff
- University of Miami Health System/Sylvester Comprehensive Cancer Center
| | | | - José Soria
- Biomedical Research Institute Sant Pau (IIB-Sant Pau)
| | | | | | | |
Collapse
|
8
|
Burdett KB, Unruh D, Drumm M, Steffens A, Lamano J, Judkins J, Schwartz M, Javier R, Amidei C, Lipp ES, Peters KB, Lai A, Eldred BSC, Heimberger AB, McCortney K, Scholtens DM, Horbinski C. Determining venous thromboembolism risk in patients with adult-type diffuse glioma. Blood 2023; 141:1322-1336. [PMID: 36399711 PMCID: PMC10082363 DOI: 10.1182/blood.2022017858] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
Venous thromboembolism (VTE) is a life-threating condition that is common in patients with adult-type diffuse gliomas, yet thromboprophylaxis is controversial because of possible intracerebral hemorrhage. Effective VTE prediction models exist for other cancers, but not glioma. Our objective was to develop a VTE prediction tool to improve glioma patient care, incorporating clinical, blood-based, histologic, and molecular markers. We analyzed preoperative arterial blood, tumor tissue, and clinical-pathologic data (including next-generation sequencing data) from 258 patients with newly diagnosed World Health Organization (WHO) grade 2 to 4 adult-type diffuse gliomas. Forty-six (17.8%) experienced VTE. Tumor expression of tissue factor (TF) and podoplanin (PDPN) each positively correlated with VTE, although only circulating TF and D-dimers, not circulating PDPN, correlated with VTE risk. Gliomas with mutations in isocitrate dehydrogenase 1 (IDH1) or IDH2 (IDHmut) caused fewer VTEs; multivariable analysis suggested that this is due to IDHmut suppression of TF, not PDPN. In a predictive time-to-event model, the following predicted increased VTE risk in newly diagnosed patients with glioma: (1) history of VTE; (2) hypertension; (3) asthma; (4) white blood cell count; (5) WHO tumor grade; (6) patient age; and (7) body mass index. Conversely, IDHmut, hypothyroidism, and MGMT promoter methylation predicted reduced VTE risk. These 10 variables were used to create a web-based VTE prediction tool that was validated in 2 separate cohorts of patients with adult-type diffuse glioma from other institutions. This study extends our understanding of the VTE landscape in these tumors and provides evidence-based guidance for clinicians to mitigate VTE risk in patients with glioma.
Collapse
Affiliation(s)
| | | | - Michael Drumm
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Alicia Steffens
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Jonathan Lamano
- Department of Neurosurgery, Stanford University, Stanford, CA
| | - Jonathan Judkins
- Department of Medicine, Oregon Health and Science University, Portland, OR
| | - Margaret Schwartz
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Rodrigo Javier
- University of Chicago Pritzker School of Medicine, Chicago, IL
| | - Christina Amidei
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Eric S. Lipp
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - Katherine B. Peters
- The Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, NC
| | - Albert Lai
- Department of Neurology, University of California, Los Angeles, CA
| | | | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Lou and Jean Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
- Department of Pathology, Northwestern University, Chicago, IL
| |
Collapse
|
9
|
Huang Y, Lu M, Wang Y, Zhang C, Cao Y, Zhang X. Podoplanin: A potential therapeutic target for thrombotic diseases. Front Neurol 2023; 14:1118843. [PMID: 36970507 PMCID: PMC10033871 DOI: 10.3389/fneur.2023.1118843] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
As a specific lymphatic marker and a key ligand of C-type lectin-like receptor 2 (CLEC-2), podoplanin (Pdpn) is involved in various physiological and pathological processes such as growth and development, respiration, blood coagulation, lymphangiogenesis, angiogenesis, and inflammation. Thrombotic diseases constitute a major cause of disability and mortality in adults, in which thrombosis and inflammation play a crucial role. Recently, increasing evidence demonstrates the distribution and function of this glycoprotein in thrombotic diseases such as atherosclerosis, ischemic stroke, venous thrombosis, ischemic-reperfusion injury (IRI) of kidney and liver, and myocardial infarction. Evidence showed that after ischemia, Pdpn can be acquired over time by a heterogeneous cell population, which may not express Pdpn in normal conditions. In this review, the research progresses in understanding the roles and mechanisms of podoplanin in thromobotic diseases are summarized. The challenges of podoplanin-targeted approaches for disease prognosis and preventions are also discussed.
Collapse
Affiliation(s)
- Yaqian Huang
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Manli Lu
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Wang
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chunyuan Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongjun Cao
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xia Zhang
- Department of Neurology, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
10
|
Ando K, Natsumeda M, Kawamura M, Shirakawa K, Okada M, Tsukamoto Y, Eda T, Watanabe J, Saito S, Takahashi H, Kakita A, Oishi M, Fujii Y. Elevated ratio of C-type lectin-like receptor 2 level and platelet count (C2PAC) aids in the diagnosis of post-operative venous thromboembolism in IDH-wildtype gliomas. Thromb Res 2023; 223:36-43. [PMID: 36706720 DOI: 10.1016/j.thromres.2023.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/08/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Podoplanin (PDPN) is known to induce platelet aggregation via interacting with the C-type lectin-like receptor-2 on platelets and is involved in postoperative venous thromboembolism (VTE) formation. In this study, we investigate the correlation between soluble C-type lectin-like receptor (sCLEC-2) levels and PDPN expression in patients with high grade gliomas and the relationship between sCLEC-2 levels and the occurrence of VTE. MATERIALS AND METHODS Forty-four patients harboring high grade gliomas, treated surgically at the Department of Neurosurgery, Niigata University from April 2018 to August 2020, were included. Patients with high grade gliomas were divided into isocitrate dehydrogenase (IDH)- wildtype and mutant groups, and the presence or absence of VTE and the intensity of PDPN by immunohistochemistry were confirmed. Platelet counts, as well as plasma sCLEC-2 and PDPN were measured in these patients. Furthermore, the levels of sCLEC-2 concentration were divided by the platelet count (C2PAC index) for comparison. RESULTS IDH-wildtype glioma patients highly expressed PDPN (P < 0.001) compared to IDH-mutant glioma patients. In total, 9 (20.5 %) patients were diagnosed with VTE during the follow-up period, of which 8 patients harbored IDH-wildtype gliomas, and one patient an IDH-mutant glioma. Mean sCLEC-2 levels and C2PAC index in patients with IDH-wildtype gliomas were significantly higher than that of low or no PDPN expression group, which included patients with IDH-mutant gliomas (P = 0.0004, P = 0.0002). In patients with IDH-wildtype gliomas, the C2PAC index in patients with VTE was significantly higher than in patients without VTE (P = 0.0492). The optimal cutoff point of C2PAC for predicting VTE in IDH-wildtype glioma patients was 3.7 with a sensitivity of 87.5 % and specificity of 51.9 %. CONCLUSION Platelet activation is strongly involved in the development of VTE in patients with IDH-wildtype high grade gliomas, and C2PAC index is a potential marker to detect VTE formation after surgery.
Collapse
Affiliation(s)
- Kazuhiro Ando
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahide Kawamura
- Department Research and Development, LSI Medience Corporation, Tokyo, Japan
| | - Kamon Shirakawa
- Department Research and Development, LSI Medience Corporation, Tokyo, Japan
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takeyoshi Eda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Jun Watanabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shoji Saito
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Haruhiko Takahashi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
11
|
Diaz M, Schiff D. Vascular complications in patients with brain tumors. Curr Opin Oncol 2022; 34:698-704. [PMID: 35788556 DOI: 10.1097/cco.0000000000000875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Venous thromboembolism (VTE) and other vascular events are common in patients with brain tumors, but their optimal management is not firmly established, in large part due to the competing risk of intracranial hemorrhage (ICH) in this population. RECENT FINDINGS There is conflicting evidence on whether therapeutic anticoagulation increases the risk of ICH in patients with brain tumors, with several metanalysis and retrospective cohort studies showing an increased risk and others showing no differences. Current guidelines recommend anticoagulating brain tumors patients with VTE with either low-molecular weight heparin (LMWH) or direct oral anticoagulants (DOACs), and several retrospective studies have shown the risk of ICH with DOACs is similar or smaller than with LMWH. SUMMARY An increased risk of VTE exists in a variety of brain tumor types. Most patients with brain tumors and VTE should receive therapeutic anticoagulation, and recent retrospective evidence supports the use of both LMWH and DOACs as effective and relatively safe in this setting. Patients with brain tumors are also at increased risk of other vascular tumor- or treatment-related complications whose optimal management is unclear.
Collapse
Affiliation(s)
- Maria Diaz
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - David Schiff
- Division of Neuro-Oncology, Department of Neurology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
12
|
Wang Y, Peng D, Huang Y, Cao Y, Li H, Zhang X. Podoplanin: Its roles and functions in neurological diseases and brain cancers. Front Pharmacol 2022; 13:964973. [PMID: 36176432 PMCID: PMC9514838 DOI: 10.3389/fphar.2022.964973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Podoplanin is a small mucin-like glycoprotein involved in several physiological and pathological processes in the brain including development, angiogenesis, tumors, ischemic stroke and other neurological disorders. Podoplanin expression is upregulated in different cell types including choroid plexus epithelial cells, glial cells, as well as periphery infiltrated immune cells during brain development and neurological disorders. As a transmembrane protein, podoplanin interacts with other molecules in the same or neighboring cells. In the past, a lot of studies reported a pleiotropic role of podoplanin in the modulation of thrombosis, inflammation, lymphangiogenesis, angiogenesis, immune surveillance, epithelial mesenchymal transition, as well as extracellular matrix remodeling in periphery, which have been well summarized and discussed. Recently, mounting evidence demonstrates the distribution and function of this molecule in brain development and neurological disorders. In this review, we summarize the research progresses in understanding the roles and mechanisms of podoplanin in the development and disorders of the nervous system. The challenges of podoplanin-targeted approaches for disease prognosis and preventions are also discussed.
Collapse
Affiliation(s)
- Yi Wang
- Department of Neurology, The Second Affiliated Hospital of Soochow University and Clinical Research Center of Neurological Disease, Suzhou, China
| | - Dan Peng
- Department of Neurology, The Second Affiliated Hospital of Soochow University and Clinical Research Center of Neurological Disease, Suzhou, China
| | - Yaqian Huang
- Department of Neurology, The Second Affiliated Hospital of Soochow University and Clinical Research Center of Neurological Disease, Suzhou, China
| | - Yongjun Cao
- Department of Neurology, The Second Affiliated Hospital of Soochow University and Clinical Research Center of Neurological Disease, Suzhou, China
| | - Hui Li
- Department of Cardiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Hui Li, ; Xia Zhang,
| | - Xia Zhang
- Department of Neurology, The Second Affiliated Hospital of Soochow University and Clinical Research Center of Neurological Disease, Suzhou, China
- *Correspondence: Hui Li, ; Xia Zhang,
| |
Collapse
|
13
|
Mantha S, Rak J. Cancer genetic alterations and risk of venous thromboembolism. Thromb Res 2022; 213 Suppl 1:S29-S34. [DOI: 10.1016/j.thromres.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 10/18/2022]
|
14
|
Tamura R, Yoshihara K, Enomoto T. Therapeutic Strategies Focused on Cancer-Associated Hypercoagulation for Ovarian Clear Cell Carcinoma. Cancers (Basel) 2022; 14:2125. [PMID: 35565252 PMCID: PMC9099459 DOI: 10.3390/cancers14092125] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is associated with chemotherapy resistance and poor prognosis, especially in advanced cases. Although comprehensive genomic analyses have clarified the significance of genomic alterations such as ARID1A and PIK3CA mutations in OCCC, therapeutic strategies based on genomic alterations have not been confirmed. On the other hand, OCCC is clinically characterized by a high incidence of thromboembolism. Moreover, OCCC specifically shows high expression of tissue factor and interleukin-6, which play a critical role in cancer-associated hypercoagulation and may be induced by OCCC-specific genetic alterations or the endometriosis-related tumor microenvironment. In this review, we focused on the association between cancer-associated hypercoagulation and molecular biology in OCCC. Moreover, we reviewed the effectiveness of candidate drugs targeting hypercoagulation, such as tissue factor- or interleukin-6-targeting drugs, anti-inflammatory drugs, anti-hypoxia signaling drugs, anticoagulants, and combined immunotherapy with these drugs for OCCC. This review is expected to contribute to novel basic research and clinical trials for the prevention, early detection, and treatment of OCCC focused on hypercoagulation.
Collapse
Affiliation(s)
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; (R.T.); (T.E.)
| | | |
Collapse
|
15
|
Huang Y, Ding H, Luo M, Li S, Xie C, Zhong Y, Li Z. Combined analysis of clinical and laboratory markers to predict the risk of venous thromboembolism in patients with IDH1 wild-type glioblastoma. Support Care Cancer 2022; 30:6063-6069. [PMID: 35419733 DOI: 10.1007/s00520-022-07050-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE We have combined analysis of clinical and laboratory markers to try to find an optimal model to predict venous thromboembolism in isocitrate dehydrogenase 1 (IDH1) wild-type glioblastoma (GBM) by a prospective research. METHODS Patients with newly histologically confirmed IDH1 wild-type (IDH1wt) GBM were recruited for this study. Status of IDH1, PTEN, P53, BRAF, MGMT promoter methylation (MGMTp), and TERT promoter (TERTp) was determined using genetic sequencing through polymerase chain reaction (PCR). Amplification of EGFR was established through fluorescence in situ hybridization (FISH). Competing risk regression model was performed to calculate the risk of VTE. Clinical and laboratory parameters that were independently predicted risk of VTE were used to develop a risk assessment model (RAM). RESULTS One hundred thirty-one patients with IDH1wt GBM were included in the present analysis. A total of 48/131 patients (36.6%) developed VTE. D-dimer, ECOG score, and EGFR amplification were suggested to be significantly associated with the VTE risk in multivariable analysis. High ECOG score (>2), high D-dimer (>1.6 μg/ml), and EGFR amplification were used as the strongest independent predictors of increased risk of VTE. The cumulative incidence of VTE was 17.2% for patients with score 0 (n =29), 23.6% for patients with score 1 (n =55), and 63.8% for patients with score 2 (n = 35) or score 3 (n = 12) by application of a RAM. CONCLUSIONS In IDH1wt GBM patients, by applying a VTE risk assessment model, we could identify patients with a very high and low risk of VTE.
Collapse
Affiliation(s)
- Yong Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Shuiguohu street, 168 Donghu road, Wuhan, Hubei, China.,Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China
| | - Haixia Ding
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Shuiguohu street, 168 Donghu road, Wuhan, Hubei, China.,Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China
| | - Min Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Shuiguohu street, 168 Donghu road, Wuhan, Hubei, China.,Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China
| | - Sirui Li
- Department of Radiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Shuiguohu street, 168 Donghu road, Wuhan, Hubei, China.,Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China
| | - Yahua Zhong
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Shuiguohu street, 168 Donghu road, Wuhan, Hubei, China. .,Hubei Cancer Clinical Study Center, Wuhan University, Wuhan, Hubei, China.
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
16
|
Diaz M, Jo J. Venous Thrombotic Events and Anticoagulation in Brain Tumor Patients. Curr Oncol Rep 2022; 24:493-500. [PMID: 35179708 DOI: 10.1007/s11912-021-01178-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Brain tumor patients have a 20-30% risk of venous thromboembolism (VTE), with management complicated by risk of intracranial hemorrhage (ICH). Here we review the epidemiology, pathogenesis, and recommended management of VTE in brain tumors. RECENT FINDINGS New risk factors and molecular mechanisms of VTE in brain tumor patients have emerged, including the protective effect of IDH mutation in gliomas and the potential role of podoplanin-mediated platelet aggregation in thrombogenesis in these tumors. Recent studies show that the risk of ICH is not significantly higher in brain tumor patients receiving anticoagulation. Based on systemic cancer trials, direct oral anticoagulants (DOACs) may be a suitable alternative to traditional heparin treatment, but the applicability of these findings to brain tumors is unclear. Anticoagulation is indicated in the treatment of VTE for brain tumor patients, and appears to be reasonably safe; based on retrospective evidence, DOACs may be a reasonable agent.
Collapse
Affiliation(s)
- Maria Diaz
- Department of Neurology, Neurology Department, Memorial Sloan Kettering Cancer Center, 1275 York Avenue7th floor, New York, NY, C-71610065, USA
| | - Jasmin Jo
- Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University, Brody 3E137, 600 Moye Blvd, NC, 27834, Greenville, USA.
| |
Collapse
|
17
|
Abstract
Cancer-associated thrombosis (including venous thromboembolism (VTE) and arterial events) is highly consequential for patients with cancer and is associated with worsened survival. Despite substantial improvements in cancer treatment, the risk of VTE has increased in recent years; VTE rates additionally depend on the type of cancer (with pancreas, stomach and primary brain tumours having the highest risk) as well as on individual patient's and cancer treatment factors. Multiple cancer-specific mechanisms of VTE have been identified and can be classified as mechanisms in which the tumour expresses proteins that alter host systems, such as levels of platelets and leukocytes, and in which the tumour expresses procoagulant proteins released into the circulation that directly activate the coagulation cascade or platelets, such as tissue factor and podoplanin, respectively. As signs and symptoms of VTE may be non-specific, diagnosis requires clinical assessment, evaluation of pre-test probability, and objective diagnostic testing with ultrasonography or CT. Risk assessment tools have been validated to identify patients at risk of VTE. Primary prevention of VTE (thromboprophylaxis) has long been recommended in the inpatient and post-surgical settings, and is now an option in the outpatient setting for individuals with high-risk cancer. Anticoagulant therapy is the cornerstone of therapy, with low molecular weight heparin or newer options such as direct oral anticoagulants. Personalized treatment incorporating risk of bleeding and patient preferences is essential, especially as a diagnosis of VTE is often considered by patients even more distressing than their cancer diagnosis, and can severely affect the quality of life. Future research should focus on current knowledge gaps including optimizing risk assessment tools, biomarker discovery, next-generation anticoagulant development and implementation science.
Collapse
|
18
|
Impacto del perfil mutacional sobre el riesgo trombótico en pacientes con cáncer. Rev Clin Esp 2022. [DOI: 10.1016/j.rce.2021.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
West MT, Kartika T, Paquin AR, Liederbauer E, Zheng TJ, Lane L, Thein K, Shatzel JJ. Thrombotic events in patients using cyclin dependent kinase 4/6 inhibitors, analysis of existing ambulatory risk assessment models and the potential influences of tumor specific risk factors. Curr Probl Cancer 2022; 46:100832. [PMID: 35034766 DOI: 10.1016/j.currproblcancer.2021.100832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022]
Abstract
Cyclin dependent kinase 4 of 6 inhibitors (CDKi) are key therapeutics in the treatment of advanced breast cancer and have recently been approved in small cell lung cancer for the prevention of myelosuppression. Thrombotic events have emerged as a significant treatment related adverse event in up to 5% of patients in clinical trials and has been reported at higher rates, up to 10%, in real world analysis. The prothrombotic mechanisms of CDKis, however, remain unknown. Cancer specific risk assessment models exist to identify who may be at highest risk of thrombosis and who could potentially benefit from prophylactic anticoagulation. However, these models may not be accurate in patients taking CDKis and may not fully capture recently identified thrombotic risk factors such as tumor specific somatic mutations. In the following manuscript, we summarize the literature on thrombotic events with CDKis in clinical trials and real-world settings, review the existing thrombosis risk assessment models for ambulatory cancer patients, and discuss the literature on tumor mutations and role in cancer associated thrombosis.
Collapse
Affiliation(s)
- Malinda T West
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon.
| | - Thomas Kartika
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Ashley R Paquin
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Erik Liederbauer
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Tony J Zheng
- OHSU School of Medicine, Department of Medicine, Oregon Health & Science University, Portland, Oregon; OHSU School of Medicine, Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Lucy Lane
- Department of Radiology, University of Vermont, Burlington, VT
| | - Kyaw Thein
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon
| | - Joseph J Shatzel
- OHSU Knight Cancer Institute, Department of Hematology and Oncology, Oregon Health & Science University, Portland, Oregon; OHSU School of Medicine, Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
20
|
Orešković D, Kaštelančić A, Raguž M, Almahariq F, Romić D, Dlaka D, Janeš A, Milotić V, Novaković S, Chudy D. Glycemia and venous thromboembolism in patients with primary brain tumors - A speculative review. Med Hypotheses 2021; 157:110719. [PMID: 34717073 DOI: 10.1016/j.mehy.2021.110719] [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: 09/02/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023]
Abstract
Venous thromboembolism (VTE) is a significant public health issue causing severe morbidity and mortality. One of the most vulnerable populations for VTE development are cancer patients. And among them, patients with brain tumors have arguably the highest risk of developing this often fatal complication. Hyperglycemia is a well-known factor which leads to a wide variety of pro-thrombotic changes. In this article, we review the current literature on the topic of VTE in brain tumor patients. We also discuss the known correlation between VTE and glycemia, as well as the importance and frequency of glycemia dysregulation in brain tumor patients. Based on the already well-known importance of glucose metabolism in cancer patients, as well as the previous research of our group, we hypothesize that there is a significant number of brain tumor patients who have chronically elevated glycemia, a fact that so-far hasn't been reported. We argue that these patients carry a significantly higher risk of VTE development and would benefit greatly from strict glycemic control. We present our hypothesis, the ways in which to test it, as well as the possible counter-arguments against it. Our hope is that other investigators will be inspired by our article to continue this type of research, since we consider the topic of VTE in brain tumor patients highly important and urgent, primarily due to its prevalence and severity.
Collapse
Affiliation(s)
- Darko Orešković
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia.
| | - Anđelo Kaštelančić
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Marina Raguž
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Fadi Almahariq
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Dominik Romić
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Domagoj Dlaka
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Andrea Janeš
- Department of Clinical Microbiology and Hospital Infections, Clinical Hospital "Dubrava", Zagreb, Croatia
| | - Vivian Milotić
- Department of Radiology, General Hospital Pula, Pula, Croatia; Department of Diagnostic and Intervention Radiology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Sabina Novaković
- Depatment of Haematology, University Hospital Center "Sestre Milosrdnice", Zagreb, Croatia
| | - Darko Chudy
- Department of Neurosurgery, Clinical Hospital "Dubrava", Zagreb, Croatia; Zagreb University School of Medicine, Croatia
| |
Collapse
|
21
|
Desai PV, Krepostman N, Collins M, De Sirkar S, Hinkleman A, Walsh K, Fareed J, Darki A. Neurological Complications of Pulmonary Embolism: a Literature Review. Curr Neurol Neurosci Rep 2021; 21:59. [PMID: 34669060 PMCID: PMC8526526 DOI: 10.1007/s11910-021-01145-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW The present review discusses in-depth about neurological complications following acute venous thromboembolism (VTE). RECENT FINDINGS Intracranial hemorrhage, acute ischemic cerebrovascular events, and VTE in brain tumors are described as central nervous system (CNS) complications of PE, while peripheral neuropathy and neuropathic pain are reported as peripheral nervous system (PNS) sequelae of PE. Syncope and seizure are illustrated as atypical neurological presentations of PE. Mounting evidence suggests higher risk of venous thromboembolism (VTE) in patients with neurological diseases, but data on reverse, i.e., neurological sequelae following VTE, is underexplored. The present review is an attempt to explore some of the latter issues categorized into CNS, PNS, and atypical complications following VTE.
Collapse
Affiliation(s)
- Parth V Desai
- Department of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Nicolas Krepostman
- Departmet of Internal Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Matthew Collins
- Departmet of Internal Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Sovik De Sirkar
- Departmet of Internal Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Alexa Hinkleman
- Departmet of Internal Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Kevin Walsh
- Departmet of Internal Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Jawed Fareed
- Department of Pathology and Laboratory Medicine and Department of Pharmacology and Neuroscience, Health Science Division, Cardiovascular Research Institute, Hemostasis and Thrombosis Research Division, Loyola University, Maywood, IL, 60153, USA
| | - Amir Darki
- Department of Cardiovascular Medicine, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
22
|
Páramo JA, Marcos-Jubilar M, Lecumberri R. Impact of the mutation profile on thrombotic risk in cancer patients. Rev Clin Esp 2021; 222:93-99. [PMID: 34548256 DOI: 10.1016/j.rceng.2021.04.008] [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: 01/05/2021] [Accepted: 04/16/2021] [Indexed: 10/20/2022]
Abstract
Patients with cancer present with an elevated risk of thrombosis, which entails high morbidity and mortality. Various predictive scales that incorporate clinical and biological data have been developed to identify those at high risk of thrombosis, but, in general, they do not allow for the optimal selection of subjects who are candidates for thromboprophylaxis. Recent studies have demonstrated that the mutation profile has a high impact on the risk of thrombosis; this will facilitate developing new predictive models of thrombosis in patients with cancer.
Collapse
Affiliation(s)
- J A Páramo
- Servicio de Hematología, Clínica Universidad de Navarra, Pamplona, Spain.
| | - M Marcos-Jubilar
- Servicio de Hematología, Clínica Universidad de Navarra, Pamplona, Spain
| | - R Lecumberri
- Servicio de Hematología, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
23
|
Venous thromboembolism incidence in cancer patients with germline BRCA mutations. Clin Transl Oncol 2021; 24:154-158. [PMID: 34374030 DOI: 10.1007/s12094-021-02678-7] [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: 03/09/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Germline BRCA (gBRCA) mutations predispose to an increased risk of breast and ovarian cancer among other neoplasms. Recently, several genomic alterations such as ALK and ROS-1 rearrangements have been described as molecular drivers of venous thromboembolism (VTE). The association of gBRCA mutations and VTE is unknown. MATERIALS AND METHODS We performed an observational, retrospective, single-center study to determine the VTE incidence in consecutive patients with gBRCA mutations and cancer diagnosis attended in the multidisciplinary heredofamiliar cancer unit (HFCU) of Hospital General Universitario Gregorio Marañón, Spain, from 2010 to 2019. RESULTS One-hundred and forty-one patients were included in the analysis. The overall VTE incidence was 12.8%. The highest incidence was reported in ovarian cancer patients (20.0%), followed by patients with both ovarian and breast cancers (16.6%) and the lowest was found in breast cancer (4.9%). No difference in the type of gBRCA mutation (1 or 2) in terms of VTE rate was observed. Sixty one percent of the patients were receiving anti-cancer therapy at the time of VTE diagnosis and the majority of the events (83.3%) were diagnosed in ambulatory setting. Khorana score was of limited value to detect high-risk patients. CONCLUSIONS The VTE incidence observed in our study is consistent with prior data described in general population of breast and ovarian cancer. The risk of VTE in these patients seems to be driven by the type of cancer. We have not observed any significant interaction of gBRCA mutation status and cancer-associated thrombosis.
Collapse
|
24
|
Mir Seyed Nazari P, Berghoff AS, Preusser M, Moik F, Posch F, Ricken G, Riedl J, Hell L, Marosi C, Hainfellner JA, Pabinger I, Ay C. Association of programmed cell death ligand 1 and circulating lymphocytes with risk of venous thromboembolism in patients with glioma. ESMO Open 2021; 5:e000647. [PMID: 32424065 PMCID: PMC7239522 DOI: 10.1136/esmoopen-2019-000647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/12/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction The role of the adaptive immune system in the pathophysiology of cancer-associated venous thromboembolism (VTE) has not been investigated in detail. Programmed cell death ligand 1 (PD-L1) is an immune checkpoint molecule responsible for immune evasion in several cancer entities, as expression on tumour cells silences the T cell-mediated immune response. Given the interrelation between inflammation, haemostasis and cancer, we aimed to investigate the association of players of the adaptive immunity (eg, lymphocytes, tumour PD-L1) with risk of VTE in patients with glioma, one of the most prothrombotic cancer types. Methods In this prospective observational single-centre cohort study, patients with newly diagnosed glioma or regrowth after resection were included. Primary endpoint was objectively confirmed VTE. At study inclusion, a blood draw was performed. Tumour PD-L1 expression was assessed via immunohistochemistry. Results In total, 193 patients were included. PD-L1 expression in ≥1% of tumour cells was observed in 20/193 (10.4%) glioma. In multivariable cox-regression analysis, on adjustment for age, sex and WHO grade IV, systemic lymphocyte counts were significantly associated with risk of VTE (HR per 1 G/L increase (95% CI): 1.15 (1.03 to 1.29), p=0.013). In contrast, no significant difference in risk of VTE was found regarding the PD-L1 status: the cumulative 24 months probability of VTE was 17.0% in patients with no PD-L1 and 11.8% in those with PD-L1 expressing tumours (p=0.663). Conclusion In summary, PD-L1 expression was not associated with risk of VTE. Interestingly, peripheral lymphocytes, which are key players in adaptive immunity, were linked to an increased risk of glioma-associated VTE.
Collapse
Affiliation(s)
- Pegah Mir Seyed Nazari
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Florian Moik
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Florian Posch
- Division of Oncology, Medical University of Graz, Graz, Austria
| | - Gerda Ricken
- Institute of Neurology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Julia Riedl
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Lena Hell
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Christine Marosi
- Division of Oncology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Johannes A Hainfellner
- Institute of Neurology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Ingrid Pabinger
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Division of Haematology and Haemostaseology, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria; I M Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
25
|
Gerotziafas GT, Mahé I, Lefkou E, AboElnazar E, Abdel-Razeq H, Taher A, Antic D, Elalamy I, Syrigos K, Van Dreden P. Overview of risk assessment models for venous thromboembolism in ambulatory patients with cancer. Thromb Res 2021; 191 Suppl 1:S50-S57. [PMID: 32736779 DOI: 10.1016/s0049-3848(20)30397-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
Abstract
A B S T R A C T Important progress has been made in the development of risk assessment models (RAM) for the identification of outpatients on anticancer treatment at risk of venous thromboembolism (VTE). Since the breakthrough publication of the original Khorana risk score (KRS) more than 10 years ago, a new generation of KRS-based scores have been developed, including the Vienna Cancer and Thrombosis Study, PROTECHT, CONKO, ONCOTEV, TicOnco and the CATS/MICA score. Among these the CATS/MICA score showed that a simplified score composed of only two calibrated predictors, the type of cancer and the D-dimer levels, offers a user-friendly tool for the evaluation of cancer-associated thrombosis (CAT) risk. The COMPASS-CAT score is the first that introduced a more synthetic approach of risk evaluation by combining cancer-related predictors with patient comorbidity in a score which is designed for the types of cancer frequently seen in the community (i.e. breast, lung colon or ovarian cancers) and has been externally validated in independent studies. The Throly score is registered as part of the same group as it has a similar structure to the COMPASS-CAT score and is applicable in patients with lymphoma. The incorporation of specific biomarkers of hypercoagulability to the RAM for CAT offers the possibility to perform a precision medicine approach in the prevention of CAT. The improvement of RAM for CAT with artificial intelligence methodologies and deep learning techniques is the challenge in the near future.
Collapse
Affiliation(s)
- Grigoris T Gerotziafas
- Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France; Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France; Department of Hematology and Cell Therapy, Saint Antoine Hospital, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Sorbonne University, Paris, France.
| | - Isabelle Mahé
- Internal Medicine Department, Hôpital Louis Mourier, APHP, Colombes, Inserm UMR_S1140, Université Paris-Diderot Paris7, Paris, France
| | - Eleftheria Lefkou
- Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France
| | | | - Hiqmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Ali Taher
- Department of Internal Medicine, Division of Hematology/Oncology, American University of Beirut, Lebanon
| | - Darko Antic
- Clinic for Hematology, Clinical Center Serbia, University of Belgrade, Belgrade, Serbia
| | - Ismail Elalamy
- Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France; Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Kostas Syrigos
- Oncology Unit, 3(rd) Dept of Medicine, National and Kapodistrian University of Athens, School of Medicine, "Sotiria" General Hospital, Athens, Greece
| | - Patrick Van Dreden
- Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Faculty of Medicine, Sorbonne University, Paris, France; Clinical Research Department, Diagnostica Stago, Gennevilliers, France
| |
Collapse
|
26
|
Mandel JJ, Youssef M, Yust-Katz S, Patel AJ, Jalali A, Li Z, Wu J, Ludmir EB, de Groot JF. IDH mutation status and the development of venous thromboembolism in astrocytoma patients. J Neurol Sci 2021; 427:117538. [PMID: 34146775 DOI: 10.1016/j.jns.2021.117538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Venous thromboembolism (VTE) is a very common adverse event for astrocytoma patients, but validation of proposed risk biomarkers has been elusive. We examine whether the status of the isocitrate dehydrogenase (IDH) gene is a risk factor for the development of venous thromboembolism (VTE) in astrocytoma patients. METHODS We conducted a retrospective chart review of 282 astrocytoma patients enrolled in the PROACTIVE (Prospective Assessment of Correlative Biomarker) study at MD Anderson Cancer Center (MDACC) from 9/1/2000 until 12/31/2013. RESULTS We identified 282 astrocytoma patients consisting of 49 IDH mutant astrocytomas and 233 IDH wildtype astrocytomas. Glioblastoma was the initial histopathologic diagnosis in 30 (61.2%) of the IDH mutated astrocytomas compared to 227(97.4%) of the IDH wild type astrocytomas. VTE was identified in 52 (18.4%) of patients. VTE was diagnosed in 7 (14.3%) of the IDH mutated astrocytomas compared to 45(19.3%) of the IDH wild type astrocytoma s (p = 0.4094). Median time to VTE from diagnosis was 2.71 months. Median time to VTE from diagnosis was 2.6 months for IDH mutated astrocytomas compared to 3.06 months for the IDH wild type astrocytomas (p = 0.8663). CONCLUSIONS IDH gene status did not appear as a significant risk factor for the development of venous thromboembolism (VTE) in our cohort of astrocytoma patients. Further research into potential biomarkers for VTE may be warranted.
Collapse
Affiliation(s)
- Jacob J Mandel
- Baylor College of Medicine, Department of Neurology and Neurosurgery, One Baylor Plaza, MS NB302, Houston, TX 77030, United States of America
| | | | - Shlomit Yust-Katz
- Rabin Medical Center, Department of Neurosurgery, 39 Jabotinski St, Petah Tikva, Israel; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Akash J Patel
- Baylor College of Medicine, Department of Neurology and Neurosurgery, One Baylor Plaza, MS NB302, Houston, TX 77030, United States of America
| | - Ali Jalali
- Baylor College of Medicine, Department of Neurology and Neurosurgery, One Baylor Plaza, MS NB302, Houston, TX 77030, United States of America
| | - Ziyi Li
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Unit 1409, P. O. Box 301402, Houston, TX 77230-1402, United States of America
| | - Jimin Wu
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Unit 1409, P. O. Box 301402, Houston, TX 77230-1402, United States of America
| | - Ethan B Ludmir
- The University of Texas MD Anderson Cancer Center, Division of Radiation Oncology - Unit 1422, 1400 Pressler St., FCT6.5000, Houston, TX 77030, United States of America
| | - John F de Groot
- The University of Texas MD Anderson Cancer Center, Department of Neuro-Oncology, Unit 431, 1515 Holcombe Blvd, Houston, TX 77030-4009, United States of America.
| |
Collapse
|
27
|
Muster V, Gary T. Contrasts in Glioblastoma-Venous Thromboembolism versus Bleeding Risk. Cells 2021; 10:cells10061414. [PMID: 34200229 PMCID: PMC8228034 DOI: 10.3390/cells10061414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is among the tumor entities with an extreme thrombogenic potential and patients are at very high risk of developing a venous thromboembolism (VTE) over the course of the disease, with an incidence of up to 30% per year. Major efforts are currently being made to understand and gain novel insights into the underlying pathomechanisms of the development of VTE in patients with glioblastoma and to find appropriate biomarkers. Yet, patients with glioblastoma not only face a high thromboembolic risk but are also at risk of bleeding events. In the case of VTE, a therapeutic anticoagulation with low molecular weight heparin or, in the case of low bleeding risk, treatment with a direct oral anticoagulant, is recommended, according to recently published guidelines. With respect to an elevated bleeding risk in glioblastoma patients, therapeutic anticoagulation remains challenging in this patient group and prospective data for this vulnerable patient group are scarce, particularly with regard to direct oral anticoagulants.
Collapse
|
28
|
Dunbar A, Bolton KL, Devlin SM, Sanchez-Vega F, Gao J, Mones JV, Wills J, Kelly D, Farina M, Cordner KB, Park Y, Kishore S, Juluru K, Iyengar NM, Levine RL, Zehir A, Park W, Khorana AA, Soff GA, Mantha S. Genomic profiling identifies somatic mutations predicting thromboembolic risk in patients with solid tumors. Blood 2021; 137:2103-2113. [PMID: 33270827 PMCID: PMC8057259 DOI: 10.1182/blood.2020007488] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Venous thromboembolism (VTE) associated with cancer (CAT) is a well-described complication of cancer and a leading cause of death in patients with cancer. The purpose of this study was to assess potential associations of molecular signatures with CAT, including tumor-specific mutations and the presence of clonal hematopoiesis. We analyzed deep-coverage targeted DNA-sequencing data of >14 000 solid tumor samples using the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets platform to identify somatic alterations associated with VTE. End point was defined as the first instance of cancer-associated pulmonary embolism and/or proximal/distal lower extremity deep vein thrombosis. Cause-specific Cox proportional hazards regression was used, adjusting for pertinent clinical covariates. Of 11 695 evaluable individuals, 72% had metastatic disease at time of analysis. Tumor-specific mutations in KRAS (hazard ratio [HR], 1.34; 95% confidence interval (CI), 1.09-1.64; adjusted P = .08), STK11 (HR, 2.12; 95% CI, 1.55-2.89; adjusted P < .001), KEAP1 (HR, 1.84; 95% CI, 1.21-2.79; adjusted P = .07), CTNNB1 (HR, 1.73; 95% CI, 1.15-2.60; adjusted P = .09), CDKN2B (HR, 1.45; 95% CI, 1.13-1.85; adjusted P = .07), and MET (HR, 1.83; 95% CI, 1.15-2.92; adjusted P = .09) were associated with a significantly increased risk of CAT independent of tumor type. Mutations in SETD2 were associated with a decreased risk of CAT (HR, 0.35; 95% CI, 0.16-0.79; adjusted P = .09). The presence of clonal hematopoiesis was not associated with an increased VTE rate. This is the first large-scale analysis to elucidate tumor-specific genomic events associated with CAT. Somatic tumor mutations of STK11, KRAS, CTNNB1, KEAP1, CDKN2B, and MET were associated with an increased risk of VTE in patients with solid tumors. Further analysis is needed to validate these findings and identify additional molecular signatures unique to individual tumor types.
Collapse
Affiliation(s)
- Andrew Dunbar
- Division of Hematologic Malignancies, Department of Medicine
| | - Kelly L Bolton
- Division of Hematologic Malignancies, Department of Medicine
| | | | | | | | - Jodi V Mones
- Division of Hematologic Malignancies, Department of Medicine
| | | | | | | | | | | | | | | | - Neil M Iyengar
- Division of Solid Tumor Oncology, Department of Medicine, and
| | - Ross L Levine
- Division of Hematologic Malignancies, Department of Medicine
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Wungki Park
- Division of Solid Tumor Oncology, Department of Medicine, and
| | - Alok A Khorana
- Department of Hematology and Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Gerald A Soff
- Division of Hematologic Malignancies, Department of Medicine
| | - Simon Mantha
- Division of Hematologic Malignancies, Department of Medicine
| |
Collapse
|
29
|
Diaz M, Jo J, Smolkin M, Ratcliffe SJ, Schiff D. Risk of Venous Thromboembolism in Grade II-IV Gliomas as a Function of Molecular Subtype. Neurology 2020; 96:e1063-e1069. [PMID: 33361259 DOI: 10.1212/wnl.0000000000011414] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine the incidence of venous thromboembolism (VTE) in lower-grade gliomas (LGGs, WHO grades II-III) and to stratify the risk of VTE by molecular subtype in gliomas grade II-IV, we performed a retrospective review of a large cohort of patients with glioma. METHODS We performed a retrospective analysis of a cohort of 635 adult patients with glioma with molecular testing seen at the University of Virginia with a diagnosis of diffuse glioma established from January 2005 to August 2017. Estimates of cumulative incidence of VTE were calculated with death as competing risk; significance was determined using the Fine and Gray model. RESULTS Of 256 patients with LGG, 81 were isocitrate dehydrogenase (IDH) wild-type; 113 IDH mutant, 1p/19q codeleted; and 62 IDH mutant, 1p/19q intact. With a median follow-up of 17.9 months, the overall cumulative incidence of VTE was 8.2% for grade II (147 patients), 9.2% for grade III (109 patients), and 30.5% for grade IV (334 patients). In grade II-IV patients, absence of an IDH mutation was associated with a threefold increase in VTE risk when compared to IDH-mutant patients (hazard ratio 3.06, 95% confidence interval 2.03-4.64). In patients with glioblastoma, there was no difference in VTE incidence according to O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status. CONCLUSION Patients with LGG have a higher VTE risk compared to the general population, which is decreased, but not eliminated, in the presence of an IDH mutation. MGMT promoter methylation in glioblastoma does not affect the incidence of VTE.
Collapse
Affiliation(s)
- Maria Diaz
- From the Division of NeuroOncology (D.S.), Department of Neurology (M.D., J.J.), and Department of Public Health Sciences, Division of Biostatistics (M.S., S.J.R.), University of Virginia, Charlottesville. M.D. is currently affiliated with the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY. J.J. is currently affiliated with the Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC.
| | - Jasmin Jo
- From the Division of NeuroOncology (D.S.), Department of Neurology (M.D., J.J.), and Department of Public Health Sciences, Division of Biostatistics (M.S., S.J.R.), University of Virginia, Charlottesville. M.D. is currently affiliated with the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY. J.J. is currently affiliated with the Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC
| | - Mark Smolkin
- From the Division of NeuroOncology (D.S.), Department of Neurology (M.D., J.J.), and Department of Public Health Sciences, Division of Biostatistics (M.S., S.J.R.), University of Virginia, Charlottesville. M.D. is currently affiliated with the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY. J.J. is currently affiliated with the Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC
| | - Sarah Jane Ratcliffe
- From the Division of NeuroOncology (D.S.), Department of Neurology (M.D., J.J.), and Department of Public Health Sciences, Division of Biostatistics (M.S., S.J.R.), University of Virginia, Charlottesville. M.D. is currently affiliated with the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY. J.J. is currently affiliated with the Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC
| | - David Schiff
- From the Division of NeuroOncology (D.S.), Department of Neurology (M.D., J.J.), and Department of Public Health Sciences, Division of Biostatistics (M.S., S.J.R.), University of Virginia, Charlottesville. M.D. is currently affiliated with the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY. J.J. is currently affiliated with the Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC
| |
Collapse
|
30
|
Leiva O, Newcomb R, Connors JM, Al-Samkari H. Cancer and thrombosis: new insights to an old problem. JOURNAL DE MÉDECINE VASCULAIRE 2020; 45:6S8-6S16. [PMID: 33276943 DOI: 10.1016/s2542-4513(20)30514-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Venous thromboembolism (VTE) is a common complication in patients with cancer and portends a poor prognosis. Our understanding of the underlying pathophysiology of VTE in cancer has advanced since Trousseau first described hypercoagulability in patients with malignancy and Virchow described his famous triad of thrombosis formation. Malignancy itself induces a thrombophilic state by increasing the risk of venous stasis, endothelial injury and an imbalance of pro and anti-thrombotic factors leading to a hypercoaguable state. Additional insults to this thrombotic balance are introduced by patient-specific, treatment related and tumor-specific factors. The importance of understanding the factors associated with increased thrombosis in cancer is paramount in order to adequately identify patients who will benefit from thromboprophylaxis.
Collapse
Affiliation(s)
- O Leiva
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - R Newcomb
- Division of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - J M Connors
- Hematology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - H Al-Samkari
- Division of Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
31
|
Mechanisms and biomarkers of cancer-associated thrombosis. Transl Res 2020; 225:33-53. [PMID: 32645431 PMCID: PMC8020882 DOI: 10.1016/j.trsl.2020.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer-associated thrombosis is a leading cause of non-cancer death in cancer patients and is comprised of both arterial and venous thromboembolism (VTE). There are multiple risk factors for developing VTE, including cancer type, stage, treatment, and other medical comorbidities, which suggests that the etiology of thrombosis is multifactorial. While cancer-associated thrombosis can be treated with anticoagulation, benefits of therapy must be balanced with the increased bleeding risks seen in patients with cancer. Although risk models exist for primary and recurrent VTE, additional predictors are needed to improve model performance and discrimination of high-risk patients. This review will outline the diverse mechanisms driving thrombosis in cancer patients, as well as provide an overview of biomarkers studied in thrombosis risk and important considerations when selecting candidate biomarkers.
Collapse
|
32
|
Leiva O, Connors JM, Al-Samkari H. Impact of Tumor Genomic Mutations on Thrombotic Risk in Cancer Patients. Cancers (Basel) 2020; 12:cancers12071958. [PMID: 32707653 PMCID: PMC7409200 DOI: 10.3390/cancers12071958] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
Venous thromboembolism (VTE) is common in patients with cancer and is an important contributor to morbidity and mortality in these patients. Early thromboprophylaxis initiated only in those cancer patients at highest risk for VTE would be optimal. Risk stratification scores incorporating tumor location, laboratory values and patient characteristics have attempted to identify those patients most likely to benefit from thromboprophylaxis but even well-validated scores are not able to reliably distinguish the highest-risk patients. Recognizing that tumor genetics affect the biology and behavior of malignancies, recent studies have explored the impact of specific molecular aberrations on the rate of VTE in cancer patients. The presence of certain molecular aberrations in a variety of different cancers, including lung, colon, brain and hematologic tumors, have been associated with an increased risk of VTE and arterial thrombotic events. This review examines the findings of these studies and discusses the implications of these findings on decisions relating to thromboprophylaxis use in the clinical setting. Ultimately, the integration of tumor molecular genomic information into clinical VTE risk stratification scores in cancer patients may prove to be a major advancement in the prevention of cancer-associated thrombosis.
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02215, USA;
- Harvard Medical School, Boston, MA 02215, USA;
| | - Jean M. Connors
- Harvard Medical School, Boston, MA 02215, USA;
- Hematology Division, Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Hanny Al-Samkari
- Harvard Medical School, Boston, MA 02215, USA;
- Division of Hematology Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence: ; Tel.: +1-617-643-6214
| |
Collapse
|
33
|
Buijs JT, Versteeg HH. Genes and proteins associated with the risk for cancer-associated thrombosis. Thromb Res 2020; 191 Suppl 1:S43-S49. [DOI: 10.1016/s0049-3848(20)30396-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/21/2022]
|
34
|
Moik F, Ay C, Pabinger I. Risk prediction for cancer-associated thrombosis in ambulatory patients with cancer: past, present and future. Thromb Res 2020; 191 Suppl 1:S3-S11. [DOI: 10.1016/s0049-3848(20)30389-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/14/2019] [Accepted: 12/23/2019] [Indexed: 01/29/2023]
|
35
|
Muster V, Gary T. Incidence, Therapy, and Bleeding Risk-Cancer- Associated Thrombosis in Patients with Glioblastoma. Cancers (Basel) 2020; 12:E1354. [PMID: 32466430 PMCID: PMC7353056 DOI: 10.3390/cancers12061354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/21/2022] Open
Abstract
Cancer is an independent risk factor for the development of venous thromboembolism (VTE). Glioblastomas are amongst cancer types with the most thrombogenic potential and patients are at a particularly high risk of VTE with an incidence up to 20-30% per year. Currently, major efforts are underway to gain novel insights into risk factors and pathomechanisms to provide a better understanding of development of VTE in patients with primary brain tumors. Treatment of VTE requires therapeutic anticoagulation, which accordingly to recently-published guidelines should be performed using low molecular weight heparin or, in case of low bleeding risk, using a direct oral anticoagulant. However, this can be very challenging due to an increased risk of intracranial hemorrhage in this patient group. Furthermore, limited data are available on the subgroup of patients with primary brain tumors.
Collapse
Affiliation(s)
- Viktoria Muster
- Division of Vascular Medicine, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | | |
Collapse
|
36
|
Alan O, Telli TA, Tuylu TB, Arikan R, Demircan NC, Ercelep O, Kaya S, Babacan NA, Atasoy BM, Bozkurt S, Bayri Y, Gul D, Ekinci G, Ziyal I, Dane F, Yumuk PF. Prognostic factors in progressive high-grade glial tumors treated with systemic approach: A single center experience. J Oncol Pharm Pract 2020; 27:329-339. [PMID: 32349641 DOI: 10.1177/1078155220920684] [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] [Indexed: 11/17/2022]
Abstract
PURPOSE Malignant high-grade gliomas are the most common and aggressive type of primary brain tumor, and the prognosis is generally extremely poor. In this retrospective study, we analyzed the outcome of systemic treatment in recurrent high-grade glioma patients and the impact of prognostic factors on survivals. METHODS Data from 114 patients with recurrent high-grade glioma who received systemic treatment and followed in our clinic between 2012 and 2018 were retrospectively analyzed. Eastern Cooperative Oncology Group (ECOG) performance status, age, gender, histology, type of surgical resection, side effects after systemic treatment (deep vein thrombosis, hypertension, proteinuria), IDH1 and alpha thalassemia/mental retardation syndrome X-linked (ATRX) mutation status were investigated as prognostic factors for progression-free survival and overall survival. RESULTS At the time of diagnosis, the median age was 48 (17-77) and 68% of the patients were male. Most common pathologic subtype was glioblastoma multiforme (68%). Median follow-up duration was 9.1 months (1-68 months). Median progression-free survival and overall survival were 6.2 months and 8 months, respectively. In multivariate analysis, ECOG PS, deep venous thrombosis and the presence of ATRX and IDH1 mutation were found to be independent prognostic factors for progression-free survival (p < 0.05) and, ECOG PS, the presence of ATRX and IDH1 mutation for overall survival (p < 0.05). CONCLUSION Our study is real life data and the median progression-free survival and overall survival rates are similar to the literature. We have found ECOG PS, presence of ATRX and IDH1 mutation to be independent prognostic factors for both progression-free survival and overall survival.
Collapse
Affiliation(s)
- Ozkan Alan
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Tugba Akin Telli
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Tugba Basoglu Tuylu
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Rukiye Arikan
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Nazım Can Demircan
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Ozlem Ercelep
- Marmara University Pendik Education and Research Hospital, Medical Oncology Clinic, Istanbul, Turkey
| | - Serap Kaya
- Marmara University Pendik Education and Research Hospital, Medical Oncology Clinic, Istanbul, Turkey
| | - Nalan Akgul Babacan
- Marmara University Pendik Education and Research Hospital, Medical Oncology Clinic, Istanbul, Turkey
| | - Beste M Atasoy
- Department of Radiation Oncology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Suheyla Bozkurt
- Department of Pathology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Yasar Bayri
- Department of Neurosurgery, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Dilek Gul
- Marmara University Pendik Education and Research Hospital, Radiation Oncology Clinic, Istanbul, Turkey
| | - Gazanfer Ekinci
- Department of Radiology, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Ibrahim Ziyal
- Department of Neurosurgery, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - Faysal Dane
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| | - P Fulden Yumuk
- Division of Medical Oncology, Department of Internal Medicine, Marmara University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
37
|
Mir Seyed Nazari P, Marosi C, Moik F, Riedl J, Özer Ö, Berghoff AS, Preusser M, Hainfellner JA, Pabinger I, Zlabinger GJ, Ay C. Low Systemic Levels of Chemokine C-C Motif Ligand 3 (CCL3) are Associated with a High Risk of Venous Thromboembolism in Patients with Glioma. Cancers (Basel) 2019; 11:cancers11122020. [PMID: 31847343 PMCID: PMC6966639 DOI: 10.3390/cancers11122020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/05/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
A tight interplay between inflammation and hemostasis has been described as a potential driver for developing venous thromboembolism (VTE). Here, we investigated the association of systemic cytokine levels and risk of VTE in patients with glioma. This analysis was conducted within the prospective, observational Vienna Cancer and Thrombosis Study. Patients with glioma were included at time of diagnosis or progression and were observed for a maximum of two years. Primary endpoint was objectively confirmed VTE. At study entry, a single blood draw was performed. A panel of nine cytokines was measured in serum samples with the xMAP technology developed by Luminex. Results: Overall, 76 glioma patients were included in this analysis, and 10 (13.2%) of them developed VTE during the follow-up. Chemokine C-C motif ligand 3 (CCL3) levels were inversely associated with risk of VTE (hazard ratio [HR] per double increase, 95% confidence interval [CI]: 0.385, 95% CI: 0.161–0.925, p = 0.033), while there was no association between the risk of VTE and serum levels of interleukin (IL)-1β, IL-4, IL-6, IL-8, IL-10, IL-11, tumor necrosis factor (TNF)-α and vascular endothelial growth factor (VEGF), respectively. In conclusion, low serum levels of CCL3 were associated with an increased risk of VTE. CCL3 might serve as a potential biomarker to predict VTE risk in patients with glioma.
Collapse
Affiliation(s)
- Pegah Mir Seyed Nazari
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
| | - Christine Marosi
- Division of Oncology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (C.M.); (A.S.B.); (M.P.)
| | - Florian Moik
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
| | - Julia Riedl
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
| | - Öykü Özer
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
| | - Anna Sophie Berghoff
- Division of Oncology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (C.M.); (A.S.B.); (M.P.)
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (C.M.); (A.S.B.); (M.P.)
| | - Johannes A. Hainfellner
- Institute of Neurology and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ingrid Pabinger
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
| | - Gerhard J. Zlabinger
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Cihan Ay
- Division of Hematology and Hemostaseology, Department of Medicine I and Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria; (P.M.S.N.); (F.M.); (J.R.); (Ö.Ö.); (I.P.)
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
- Correspondence:
| |
Collapse
|
38
|
Watanabe J, Natsumeda M, Okada M, Kanemaru Y, Tsukamoto Y, Oishi M, Kakita A, Fujii Y. Podoplanin Expression and IDH-Wildtype Status Predict Venous Thromboembolism in Patients with High-Grade Gliomas in the Early Postoperative Period. World Neurosurg 2019; 128:e982-e988. [PMID: 31100523 DOI: 10.1016/j.wneu.2019.05.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/05/2019] [Accepted: 05/06/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Venous thromboembolism (VTE) often is encountered in patients with high-grade gliomas. The underlying mechanisms are unclear, as is the optimal prophylactic protocol. The purpose of the present study was to identify risk factors of VTE and examine the validity of early VTE detection in high-grade gliomas. METHODS We reviewed the medical records of 165 patients with newly diagnosed high-grade glioma treated at Niigata University Hospital during the years 2009 to 2016. If the serum D-dimer levels increased to 5.0 μg/mL or more, computed tomography was performed to detect VTE. Furthermore, immunohistochemistry with antibodies against podoplanin was performed on available 101 tumor tissues. RESULTS Of the 165 patients, 44 (26.7%) developed VTE. Of the 44 patients, 34 (79.5%) developed VTE within 7 days after surgery. No fatal VTE occurred and major complications secondary to anticoagulation occurred in only 2 (1.2%) patients. On multivariate analysis, lower Karnofsky Performance Scale status, podoplanin expression, and isocitrate dehydrogenase-wildtype status were independently associated with the risk of VTE (P < 0.05). CONCLUSIONS We found that most VTEs occurred early in the postoperative period and commonly in patients with lower Karnofsky Performance Scale status and isocitrate dehydrogenase-wildtype gliomas, which expressed podoplanin.
Collapse
Affiliation(s)
- Jun Watanabe
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata.
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| | - Yu Kanemaru
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| | - Yoshihiro Tsukamoto
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| | - Makoto Oishi
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata
| | - Yukihiko Fujii
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata
| |
Collapse
|
39
|
Riedl J, Ay C. Venous Thromboembolism in Brain Tumors: Risk Factors, Molecular Mechanisms, and Clinical Challenges. Semin Thromb Hemost 2019; 45:334-341. [PMID: 31041803 DOI: 10.1055/s-0039-1688493] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Venous thromboembolism (VTE) is a common complication in patients with primary brain tumors, with up to 20% of patients per year having a VTE event. Clinical risk factors for VTE include glioblastoma subtype, paresis, or surgery. Furthermore, specific factors playing a role in tumor biology were recently identified to predispose to prothrombotic risk. For instance, mutations in the isocitrate dehydrogenase 1 (IDH1) gene, which occurs in a subgroup of glioma, correlate with risk of VTE, with low incidence in patients with presence of an IDH1 mutation compared with those with IDH1 wild-type status. In addition, expression of the glycoprotein podoplanin on brain tumors was associated with both intratumoral thrombi and high risk of VTE. As podoplanin has the ability to activate platelets, a mechanistic role of podoplanin-mediated platelet activation in VTE development has been suggested. From a clinical point of view, the management of patients with primary brain tumors and VTE is challenging. Anticoagulation is required to treat patients; however, it is associated with increased risk of intracranial hemorrhage. This review focuses on describing the epidemiology, risk factors, and mechanisms of brain tumor-associated thrombosis and discusses clinical challenges in the prevention and treatment of VTE in patients with brain tumors.
Collapse
Affiliation(s)
- Julia Riedl
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
40
|
Mantia C, Zwicker JI. Anticoagulation in the Setting of Primary and Metastatic Brain Tumors. Cancer Treat Res 2019; 179:179-189. [PMID: 31317488 DOI: 10.1007/978-3-030-20315-3_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Venous thromboembolism is commonly diagnosed in patients with primary and secondary brain tumors. Anticoagulation management in the setting of brain tumors is complicated by the high background rate of spontaneous intracranial hemorrhage. Until recently, there was limited evidence to support the decision to administer therapeutic anticoagulation in the setting of brain metastases or primary brain tumors. The current evidence suggests that the safety profile of therapeutic low molecular weight heparin for the treatment of venous thromboembolism is contingent on whether the origin of brain tumor is primary (i.e., glioma) versus secondary. In patients with brain metastases, the rate of intracranial hemorrhage often exceeds 20% but is not influenced by the administration of low molecular weight heparin. In contrast, in primary brain tumors such as glioma, therapeutic anticoagulation is associated with an increased risk of intracranial hemorrhage that can negatively impact survival. This chapter reviews the underlying mechanisms contributing to thrombosis and hemorrhage in brain tumors and summarizes the current evidence and approaches in anticoagulation to treat venous thromboembolism.
Collapse
Affiliation(s)
- Charlene Mantia
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jeffrey I Zwicker
- Division of Hemostasis and Thrombosis, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
41
|
Mir Seyed Nazari P, Riedl J, Preusser M, Posch F, Thaler J, Marosi C, Birner P, Ricken G, Hainfellner JA, Pabinger I, Ay C. Combination of isocitrate dehydrogenase 1 (IDH1) mutation and podoplanin expression in brain tumors identifies patients at high or low risk of venous thromboembolism. J Thromb Haemost 2018; 16:1121-1127. [PMID: 29676036 PMCID: PMC6099350 DOI: 10.1111/jth.14129] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Indexed: 12/11/2022]
Abstract
Essentials Risk stratification for venous thromboembolism (VTE) in patients with brain tumors is challenging. Patients with IDH1 wildtype and high podoplanin expression have a 6-month VTE risk of 18.2%. Patients with IDH1 mutation and no podoplanin expression have a 6-month VTE risk of 0%. IDH1 mutation and podoplanin overexpression in primary brain tumors appear to be exclusive. SUMMARY Background Venous thromboembolism (VTE) is a frequent complication in primary brain tumor patients. Independent studies revealed that podoplanin expression in brain tumors is associated with increased VTE risk, whereas the isocitrate dehydrogenase 1 (IDH1) mutation is associated with very low VTE risk. Objectives To investigate the interrelation between intratumoral podoplanin expression and IDH1 mutation, and their mutual impact on VTE development. Patients/Methods In a prospective cohort study, intratumoral IDH1 R132H mutation and podoplanin were determined in brain tumor specimens (mainly glioma) by immunohistochemistry. The primary endpoint of the study was symptomatic VTE during a 2-year follow-up. Results All brain tumors that expressed podoplanin to a medium-high extent showed also an IDH1 wild-type status. A score based on IDH1 status and podoplanin expression levels allowed prediction of the risk of VTE. Patients with wild-type IDH1 brain tumors and high podoplanin expression had a significantly increased VTE risk compared with those with mutant IDH1 tumors and no podoplanin expression (6-month risk 18.2% vs. 0%). Conclusions IDH1 mutation and podoplanin overexpression seem to be exclusive. Although brain tumor patients with IDH1 mutation are at very low risk of VTE, the risk of VTE in patients with IDH1 wild-type tumors is strongly linked to podoplanin expression levels.
Collapse
Affiliation(s)
- P. Mir Seyed Nazari
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
| | - J. Riedl
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
| | - M. Preusser
- Clinical Division of OncologyDepartment of Medicine IComprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - F. Posch
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
- Division of OncologyDepartment of Internal MedicineMedical University of GrazGrazAustria
| | - J. Thaler
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
| | - C. Marosi
- Clinical Division of OncologyDepartment of Medicine IComprehensive Cancer CenterMedical University of ViennaViennaAustria
| | - P. Birner
- Clinical Institute of PathologyMedical University of ViennaViennaAustria
| | - G. Ricken
- Institute of NeurologyMedical University of ViennaViennaAustria
| | | | - I. Pabinger
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
| | - C. Ay
- Clinical Division of Hematology and HemostaseologyDepartment of Medicine IComprehensive Cancer CenterCenter Medical University of ViennaViennaAustria
| |
Collapse
|