1
|
Veiga VC, Peres SV, Ostolin TLVDP, Moraes FR, Belucci TR, Clara CA, Cavalcanti AB, Chaddad-Neto FEA, Batistella GNDR, Neville IS, Baeta AM, Yamada CAF. Incidence of venous thromboembolism and bleeding in patients with malignant central nervous system neoplasm: Systematic review and meta-analysis. PLoS One 2024; 19:e0304682. [PMID: 38900739 PMCID: PMC11189257 DOI: 10.1371/journal.pone.0304682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Central nervous system (CNS) malignant neoplasms may lead to venous thromboembolism (VTE) and bleeding, which result in rehospitalization, morbidity and mortality. We aimed to assess the incidence of VTE and bleeding in this population. METHODS This systematic review and meta-analysis (PROSPERO CRD42023423949) were based on a standardized search of PubMed, Virtual Health Library and Cochrane (n = 1653) in July 2023. After duplicate removal, data screening and collection were conducted by independent reviewers. The combined rates and 95% confidence intervals for the incidence of VTE and bleeding were calculated using the random effects model with double arcsine transformation. Subgroup analyses were performed based on sex, age, income, and type of tumor. Heterogeneity was calculated using Cochran's Q test and I2 statistics. Egger's test and funnel graphs were used to assess publication bias. RESULTS Only 36 studies were included, mainly retrospective cohorts (n = 30, 83.3%) from North America (n = 20). Most studies included were published in high-income countries. The sample size of studies varied between 34 and 21,384 adult patients, mostly based on gliomas (n = 30,045). For overall malignant primary CNS neoplasm, the pooled incidence was 13.68% (95%CI 9.79; 18.79) and 11.60% (95%CI 6.16; 18.41) for VTE and bleeding, respectively. The subgroup with elderly people aged 60 or over had the highest incidence of VTE (32.27% - 95%CI 14.40;53.31). The studies presented few biases, being mostly high quality. Despite some variability among the studies, we observed consistent results by performing sensitivity analysis, which highlight the robustness of our findings. CONCLUSIONS Our study showed variability in the pooled incidence for both overall events and subgroup analyses. It was highlighted that individuals over 60 years old or diagnosed with GBM had a higher pooled incidence of VTE among those with overall CNS malignancies. It is important to note that the results of this meta-analysis refer mainly to studies carried out in high-income countries. This highlights the need for additional research in Latin America, and low- and middle-income countries.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Alex M. Baeta
- BP–A Beneficência Portuguesa de São Paulo, São Paulo, Brasil
| | | | | |
Collapse
|
2
|
Rolling CC, Mohme M, Bokemeyer C, Westphal M, Riethdorf S, Lamszus K, Pantel K, Klingler F, Langer F. Circulating Tumor Cells and Thromboembolic Events in Patients with Glioblastoma. Hamostaseologie 2024. [PMID: 38636546 DOI: 10.1055/a-2251-6766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024] Open
Abstract
Patients with glioblastoma (GBM) are at increased risk for arterial and venous thromboembolism (TE). Risk factors include surgery, the use of corticosteroids, radiation, and chemotherapy, but also prothrombotic characteristics of the tumor itself such as expression of tissue factor, vascular endothelial growth factor, or podoplanin. Although distant metastases are extremely rare in this tumor entity, circulating tumor cells (CTCs) have been detected in a significant proportion of GBM patients, potentially linking local tumor growth characteristics to systemic hypercoagulability. We performed post hoc analysis of a study, in which GBM patients had been investigated for CTCs. Information on TE was retrieved from electronic patient charts. In total, 133 patients (median age, 63 years; interquartile range, 53-70 years) were analyzed. During follow-up, TE was documented in 14 patients (11%), including 8 venous and 6 arterial events. CTCs were detected in 26 patients (20%). Four (15%) patients with CTCs had a TE compared with 10 (9%) patients without CTCs. There was no difference in the frequency of TE events between patients with and those without detectable CTCs (p = 0.58). In summary, although our study confirms a high risk of TE in GBM patients, it does not point to an obvious association between CTCs and vascular thrombosis.
Collapse
Affiliation(s)
- Christina C Rolling
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Riethdorf
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumour Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Klingler
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Langer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Lim-Fat MJ, Iorgulescu JB, Rahman R, Bhave V, Muzikansky A, Woodward E, Whorral S, Allen M, Touat M, Li X, Xy G, Patel J, Gerstner ER, Kalpathy-Cramer J, Youssef G, Chukwueke U, McFaline-Figueroa JR, Nayak L, Lee EQ, Reardon DA, Beroukhim R, Huang RY, Bi WL, Ligon KL, Wen PY. Clinical and Genomic Predictors of Adverse Events in Newly Diagnosed Glioblastoma. Clin Cancer Res 2024; 30:1327-1337. [PMID: 38252427 DOI: 10.1158/1078-0432.ccr-23-3018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
PURPOSE Adverse clinical events cause significant morbidity in patients with GBM (GBM). We examined whether genomic alterations were associated with AE (AE) in patients with GBM. EXPERIMENTAL DESIGN We identified adults with histologically confirmed IDH-wild-type GBM with targeted next-generation sequencing (OncoPanel) at Dana Farber Cancer Institute from 2013 to 2019. Seizure at presentation, lymphopenia, thromboembolic events, pseudoprogression, and early progression (within 6 months of diagnosis) were identified as AE. The biologic function of genetic variants was categorized as loss-of-function (LoF), no change in function, or gain-of-function (GoF) using a somatic tumor mutation knowledge base (OncoKB) and consensus protein function predictions. Associations between functional genomic alterations and AE were examined using univariate logistic regressions and multivariable regressions adjusted for additional clinical predictors. RESULTS Our study included 470 patients diagnosed with GBM who met the study criteria. We focused on 105 genes that had sequencing data available for ≥ 90% of the patients and were altered in ≥10% of the cohort. Following false-discovery rate (FDR) correction and multivariable adjustment, the TP53, RB1, IGF1R, and DIS3 LoF alterations were associated with lower odds of seizures, while EGFR, SMARCA4, GNA11, BRD4, and TCF3 GoF and SETD2 LoF alterations were associated with higher odds of seizures. For all other AE of interest, no significant associations were found with genomic alterations following FDR correction. CONCLUSIONS Genomic biomarkers based on functional variant analysis of a routine clinical panel may help identify AE in GBM, particularly seizures. Identifying these risk factors could improve the management of patients through better supportive care and consideration of prophylactic therapies.
Collapse
Affiliation(s)
- Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - J Bryan Iorgulescu
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rifaquat Rahman
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Varun Bhave
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alona Muzikansky
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Eleanor Woodward
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sydney Whorral
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marie Allen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mehdi Touat
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | | | | | - Jay Patel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth R Gerstner
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Gilbert Youssef
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ugonma Chukwueke
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - J Ricardo McFaline-Figueroa
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rameen Beroukhim
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond Y Huang
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith L Ligon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
V Bonm A, Taylor LP. Management of Neurologic Complications in Patients with Brain and Spine Tumors. Semin Neurol 2024; 44:53-63. [PMID: 38176424 DOI: 10.1055/s-0043-1777422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
Patients with brain and spine tumors represent a distinct population with unique needs. We provide a practical review of neurologic care in this group with an emphasis on familiarizing the general neurologist to the nuances of neuro-oncologic supportive care. We review the management of cerebral edema, steroid dosing, and pertinent side effects. We discuss seizure management, including choice of anticonvulsants, putative antitumor effects, and important seizure mimics like drop attacks. We review the presentation and symptomatology of stroke-like migraine attack after radiation therapy (SMART syndrome). We describe the signs and symptoms that should prompt concern for metastatic spinal cord compression, as well as both acute and definitive treatment options. Finally, we discuss the underappreciated incidence of venous thromboembolic events, particularly in patients with gliomas, and review the data on management.
Collapse
Affiliation(s)
- Alipi V Bonm
- Department of Neurology, Swedish Neuroscience Institute, Seattle, Washington
| | - Lynne P Taylor
- Department of Neurology, University of Washington, Seattle, Washington
| |
Collapse
|
5
|
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
|
6
|
Welch MR. Management of Complications in Neuro-oncology Patients. Continuum (Minneap Minn) 2023; 29:1844-1871. [PMID: 38085901 DOI: 10.1212/con.0000000000001359] [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: 12/18/2023]
Abstract
OBJECTIVE The purpose of this article is to familiarize the reader with the spectrum of neurologic and medical complications relevant to the care of patients with neurologic cancer while highlighting best practices to prevent morbidity and mortality. Topics include tumor-related epilepsy, vasogenic edema, complications of corticosteroid use, disruption of the hypothalamic-pituitary axis, venous thromboembolism, and opportunistic infection. LATEST DEVELOPMENTS In 2021, a joint guideline from the Society for Neuro-Oncology and the European Association of Neuro-Oncology reaffirmed recommendations first established in 2000 that patients with newly diagnosed brain tumors should not be prescribed an antiseizure medication prophylactically. For those with tumor-related epilepsy, monotherapy with a non-enzyme-inducing anticonvulsant is the preferred initial treatment, and levetiracetam remains the preferred first choice. Surveys of physician practice continue to demonstrate excessive use of glucocorticoids in the management of patients with both primary and metastatic central nervous system malignancy. This is particularly concerning among patients who require checkpoint inhibitors as the efficacy of these agents is blunted by concomitant glucocorticoid use, resulting in a reduction in overall survival. Finally, direct oral anticoagulants have been shown to be safe in patients with brain tumors and are now favored as first-line treatment among those who require treatment for venous thromboembolism. ESSENTIAL POINTS Medical care for patients impacted by primary and secondary central nervous system malignancy is complex and requires a committed team-based approach that routinely calls upon the expertise of physicians across multiple fields. Neurologists have an important role to play and should be familiar with the spectrum of complications impacting these patients as well as the latest recommendations for management.
Collapse
|
7
|
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
|
8
|
Kaye B, Ali A, Correa Bastianon Santiago RA, Ibrahim B, Isidor J, Awad H, Sabahi M, Obrzut M, Adada B, Ranjan S, Borghei-Razavi H. The Role of EGFR Amplification in Deep Venous Thrombosis Occurrence in IDH Wild-Type Glioblastoma. Curr Oncol 2023; 30:4946-4956. [PMID: 37232831 DOI: 10.3390/curroncol30050373] [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: 03/21/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction: Glioblastoma (GBM) patients have a 20-30 incidence of venous thromboembolic events. EGFR is a widely used prognostic marker for many cancers. Recent lung cancer studies have described relationships between EGFR amplification and an increased incidence of thromboembolic complications. We aim to explore this relationship in glioblastoma patients. Methods: Two hundred ninety-three consecutive patients with IDH wild-type GBM were included in the analysis. The amplification status of EGFR was measured using fluorescence in situ hybridization (FISH). Centromere 7 (CEP7) expression was recorded to calculate the EGFR-to-CEP7 ratio. All data were collected retrospectively through chart review. Molecular data were obtained through the surgical pathology report at the time of biopsy. Results: There were 112 subjects who were EGFR-amplified (38.2%) and 181 who were non-amplified (61.8%). EGFR amplification status was not significantly correlated with VTE risk overall (p = 0.2001). There was no statistically significant association between VTE and EGFR status after controlling for Bevacizumab therapy (p = 0.1626). EGFR non-amplified status was associated with an increased VTE risk in subjects greater than 60 years of age (p = 0.048). Conclusions: There was no significant difference in occurrence of VTE in patients with glioblastoma, regardless of EGFR amplification status. Patients older than 60 years of age with EGFR amplification experienced a lower rate of VTE, contrary to some reports on non-small-cell lung cancer linking EGFR amplification to VTE risk.
Collapse
Affiliation(s)
- Brandon Kaye
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Assad Ali
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | | | - Bilal Ibrahim
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | - Julio Isidor
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | - Hany Awad
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | | | - Michal Obrzut
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | - Badih Adada
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | - Surabhi Ranjan
- Cleveland Clinic Florida, Department of Neurosurgery, Weston, FL 33331, USA
| | | |
Collapse
|
9
|
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
|
10
|
Miller JJ, Gonzalez Castro LN, McBrayer S, Weller M, Cloughesy T, Portnow J, Andronesi O, Barnholtz-Sloan JS, Baumert BG, Berger MS, Bi WL, Bindra R, Cahill DP, Chang SM, Costello JF, Horbinski C, Huang RY, Jenkins RB, Ligon KL, Mellinghoff IK, Nabors LB, Platten M, Reardon DA, Shi DD, Schiff D, Wick W, Yan H, von Deimling A, van den Bent M, Kaelin WG, Wen PY. Isocitrate dehydrogenase (IDH) mutant gliomas: A Society for Neuro-Oncology (SNO) consensus review on diagnosis, management, and future directions. Neuro Oncol 2023; 25:4-25. [PMID: 36239925 PMCID: PMC9825337 DOI: 10.1093/neuonc/noac207] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Isocitrate dehydrogenase (IDH) mutant gliomas are the most common adult, malignant primary brain tumors diagnosed in patients younger than 50, constituting an important cause of morbidity and mortality. In recent years, there has been significant progress in understanding the molecular pathogenesis and biology of these tumors, sparking multiple efforts to improve their diagnosis and treatment. In this consensus review from the Society for Neuro-Oncology (SNO), the current diagnosis and management of IDH-mutant gliomas will be discussed. In addition, novel therapies, such as targeted molecular therapies and immunotherapies, will be reviewed. Current challenges and future directions for research will be discussed.
Collapse
Affiliation(s)
- Julie J Miller
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - L Nicolas Gonzalez Castro
- Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Samuel McBrayer
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, Texas, 75235, USA
| | - Michael Weller
- Department of Neurology, University Hospital Zurich, Frauenklinikstrasse 26, 8091 Zurich, Switzerland
| | | | - Jana Portnow
- Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Ovidiu Andronesi
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Jill S Barnholtz-Sloan
- Informatics and Data Science (IDS), Center for Biomedical Informatics and Information Technology (CBIIT), Trans-Divisional Research Program (TDRP), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Brigitta G Baumert
- Cantonal Hospital Graubunden, Institute of Radiation-Oncology, Chur, Switzerland
| | - Mitchell S Berger
- Department of Neurosurgery, University of California-San Francisco, San Francisco, California, USA
| | - Wenya Linda Bi
- Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ranjit Bindra
- Department of Therapeutic Radiology, Brain Tumor Center, Yale School of Medicine, New Haven, CT, USA
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Susan M Chang
- Department of Neurosurgery, University of California-San Francisco, San Francisco, California, USA
| | - Joseph F Costello
- Department of Neurosurgery, University of California-San Francisco, San Francisco, California, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Raymond Y Huang
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Robert B Jenkins
- Individualized Medicine Research, Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, Minnesota 55901, USA
| | - Keith L Ligon
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Ingo K Mellinghoff
- Department of Neurology, Evnin Family Chair in Neuro-Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - L Burt Nabors
- Department of Neurology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael Platten
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - David A Reardon
- Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Diana D Shi
- Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - David Schiff
- Division of Neuro-Oncology, Department of Neurology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Wolfgang Wick
- Neuro-Oncology at the German Cancer Research Center (DKFZ), Program Chair of Neuro-Oncology at the National Center for Tumor Diseases (NCT), and Neurology and Chairman at the Neurology Clinic in Heidelberg, Heidelberg, Germany
| | - Hai Yan
- Genetron Health Inc, Gaithersburg, Maryland 20879, USA
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, and, Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), and, DKTK, INF 224, 69120 Heidelberg, Germany
| | - Martin van den Bent
- Brain Tumour Centre, Erasmus MC Cancer Institute, Groene Hilledijk 301, 3075 EA Rotterdam, The Netherlands
| | - William G Kaelin
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA
| |
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
|
Low SK, Anjum Z, Mahmoud A, Joshi U, Kouides P. Isocitrate dehydrogenase mutation and risk of venous thromboembolism in glioma: A systematic review and meta-analysis. Thromb Res 2022; 219:14-21. [PMID: 36088710 DOI: 10.1016/j.thromres.2022.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Patients with malignancies including malignant gliomas have a relatively high risk for venous thromboembolism (VTE). Recent evidence has linked isocitrate dehydrogenase (IDH) mutation with reduced VTE risk in malignant glioma. This meta-analysis aims to quantify the association of IDH mutation status with risks of VTE in patients with glioma. METHODS We searched PubMed, Google Scholar, Medline OVID, Cochrane library, Cumulative Index to Nursing and Allied Health Literature databases to identify relevant studies. The overall odd ratio (OR) was pooled using the random-effects model. We evaluated the statistical heterogeneity using Cochran's Q statistics and I2 tests. We performed subgroup analyses according to age, tumor, study design, and study quality. RESULTS A total of 2600 patients from 8 studies were included in the meta-analysis. Patients with IDH mutant-type gliomas had a significantly lower risk of VTE (OR: 0.21, 95 % confidence interval [CI]: 0.09-0.46, I2 = 34 %) compared to patients with IDH wild-type gliomas. Among high-grade (III and IV) glioma, VTE events in IDH-mutant gliomas occurred with an OR of 0.28 (95 % CI: 0.14-0.53). No statistically significant decrease in the VTE risk was observed in grade II gliomas with IDH mutation compared to IDH wild-type gliomas, as indicated by the OR of 0.60 (95 % CI: 0.17-2.11). CONCLUSION IDH mutation is significantly associated with 79 % lower risk of VTE among patients with high-grade glioma compared to IDH wild-type. Our findings suggest the potential utility of IDH mutation status regarding thromboprophylaxis, and the need for further studies to elucidate the mechanism of the association.
Collapse
Affiliation(s)
- Soon Khai Low
- Department of Internal Medicine, Rochester General Hospital, NY, United States of America.
| | - Zauraiz Anjum
- Department of Internal Medicine, Rochester General Hospital, NY, United States of America
| | - Amir Mahmoud
- Department of Internal Medicine, Rochester General Hospital, NY, United States of America
| | - Utsav Joshi
- Department of Internal Medicine, Rochester General Hospital, NY, United States of America
| | - Peter Kouides
- Department of Hematology, Lipson Cancer Institute, Rochester General Hospital, NY, United States of America
| |
Collapse
|
13
|
Eisele A, Seystahl K, Rushing EJ, Roth P, Le Rhun E, Weller M, Gramatzki D. Venous thromboembolic events in glioblastoma patients: an epidemiological study. Eur J Neurol 2022; 29:2386-2397. [PMID: 35545894 PMCID: PMC9543144 DOI: 10.1111/ene.15404] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 12/01/2022]
Abstract
Background and purpose Venous thromboembolic events (VTEs) are a major complication in cancer patients, and therefore, also in brain cancer patients, anticoagulants are considered appropriate in the treatment of VTEs. Methods Frequency, risk factors, and treatment of VTEs, as well as associated complications, were assessed in a population‐based cohort of glioblastoma patients in the Canton of Zurich, Switzerland. Correlations between clinical data and survival were retrospectively analyzed using the log‐rank test and Cox regression models. Results Four hundred fourteen glioblastoma patients with isocitrate dehydrogenase wild‐type status were identified. VTEs were documented in 65 patients (15.7%). Median time from tumor diagnosis to the occurrence of a VTE was 1.8 months, and 27 patients were diagnosed with VTEs postoperatively (within 35 days; 42.2%). History of a prior VTE was more common in patients who developed VTEs than in those who did not (p = 0.004). Bevacizumab treatment at any time during the disease course was not associated with occurrence of VTEs (p = 0.593). Most patients with VTEs (n = 61, 93.8%) were treated with therapeutic anticoagulation. Complications occurred in 14 patients (23.0%), mainly intracranial hemorrhages (n = 7, 11.5%). Overall survival did not differ between patients diagnosed with VTEs and those who had no VTE (p = 0.139). Tumor progression was the major cause of death (n = 283, 90.7%), and only three patients (1.0%) died in association with acute VTEs. Conclusions Venous thromboembolic events occurred early in the disease course, suggesting that the implementation of primary venous thromboembolism prophylaxis during first‐line chemoradiotherapy could be explored in a randomized setting.
Collapse
Affiliation(s)
- Amanda Eisele
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Katharina Seystahl
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick Roth
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emilie Le Rhun
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dorothee Gramatzki
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Taylor JW. Treat the VTE and worry less about the hemorrhage: A sigh of relief or is the jury still out for therapeutic anticoagulation in high grade glioma? Neuro Oncol 2021; 24:465-466. [PMID: 34850168 DOI: 10.1093/neuonc/noab277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jennie W Taylor
- University of California, San Francisco, Neurology and Neurological Surgery
| |
Collapse
|
17
|
Guo Y, Gao X, An S, Li X, Pan L, Liu H, Liu J, Gao J, Zhao Z, Li G, Han Y, Li Y, Ji Z. Deletion of miR-15a inhibited glioma development via targeting Smad7 and inhibiting EMT pathway. Aging (Albany NY) 2021; 13:24339-24348. [PMID: 34775378 PMCID: PMC8610134 DOI: 10.18632/aging.203684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022]
Abstract
In the present study, we found the expression of miR-15a-5p (miR-15a) was increased in glioma tissues, and we further explore the underlying mechanism of miR-15a in glioma progression. Microarray analysis used to identify the differentially expressed microRNAs (miRNAs) in glioma tissues. The expression of miR-15a in glioma tissues and cell lines was tested by qRT-PCR. Luciferase assay was used to determine the binding between miR-15a and Smad7. Wound healing and transwell assay were used to examine the role of miR-15a/Smad7 in SHG139 cells. Western blot was used to detect the protein level of Smad7 and epithelial-mesenchymal transition (EMT) markers. A tumor formation model in nude mice was established to measure the role of miR-15a in vivo. MiR-15a was significantly increased in glioma tissues and cells, which indicated a poor prognosis of glioma patients. MiR-15a mimics induced miR-15a level in SHG139 cells, and promoted the malignancy of SHG139 cells, while miR-15a inhibitor showed the opposite effects. Luciferase assay indicated that Smad7 was the direct target of miR-15a, and Smad7 was down-regulated in glioma tissues. Functional experiments revealed that miR-15a inhibitor inhibited the EMT pathway and the migration and invasion of glioma cells, but the silencing of Smad7 reversed the effects of miR-15a inhibitor in EMT pathway and glioma progression. Finally, we performed animal experiments to verify the role of miR-15a in vivo. Present study showed that deletion of miR-15a inhibited the activation of EMT signaling via targeting Smad7, thus suppressed the tumorigenesis and tumor growth of glioma.
Collapse
Affiliation(s)
- Yanfeng Guo
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Xiaopeng Gao
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Shien An
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Xin Li
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Lekun Pan
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Hongyan Liu
- Department of E.N.T, The First Hospital of Handan, Handan, Hebei Province, China
| | - Jixiang Liu
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Jianzhou Gao
- Department of Neurosurgery, The First Hospital of Handan, Handan, Hebei Province, China
| | - Zhihuang Zhao
- Third Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Gang Li
- Third Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yonggang Han
- Third Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yabin Li
- Third Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Zhisheng Ji
- Third Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| |
Collapse
|
18
|
Winther-Larsen A, Sandfeld-Paulsen B, Hvas AM. New Insights in Coagulation and Fibrinolysis in Patients with Primary Brain Cancer: A Systematic Review. Semin Thromb Hemost 2021; 48:323-337. [PMID: 34624915 DOI: 10.1055/s-0041-1733961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Patients with primary brain tumors have a high incidence of thrombosis and hemorrhage. The underlying mechanism is believed to be derangement of their hemostatic system. To get nearer a clarification of this, we aimed to systematically review the existing literature regarding primary and secondary hemostasis as well as fibrinolysis in patients with primary brain tumor. The review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The databases PubMed, Embase, and Web of Science were searched on December 15, 2020, without time restrictions. Studies were included if they evaluated at least one blood coagulation and/or fibrinolysis parameter in patients with primary brain cancer. In total, 26 articles including 3,288 patients were included. Overall, increased activity of secondary hemostasis was observed as increased prothrombin fragment 1 + 2 and endogenous thrombin generation levels were found in glioma patients compared with controls. Furthermore, data showed a state of hypofibrinolysis with increased plasminogen activator inhibitor 1 and prolonged clot lysis time in glioma patients. In contrast, no consistent increase in the primary hemostasis was identified; however, data suggested that increased sP-selectin could be a biomarker of increased venous thromboembolism risk and that increased platelet count may be prognostic for survival. Lastly, data indicated that fibrinogen and D-dimer could hold prognostic value. In conclusion, this review indicates that an increased activity of secondary hemostasis and impaired fibrinolysis could be important players in the pathogeneses behind the high risk of thromboembolisms observed in brain cancer patients. Thus, long-term thromboprophylaxis may be beneficial and additional studies addressing this issue are wanted.
Collapse
Affiliation(s)
- Anne Winther-Larsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anne-Mette Hvas
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
19
|
Jo J, Donahue J, Sarai G, Petroni G, Schiff D. Management of Venous Thromboembolism in High-Grade Glioma: Does Low Molecular Weight Heparin Increase Intracranial Bleeding Risk? Neuro Oncol 2021; 24:455-464. [PMID: 34383073 DOI: 10.1093/neuonc/noab198] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Venous thromboembolism (VTE) occurs in up to 30% of patients with high-grade glioma (HGG). Concern for increased risk of intracranial hemorrhage (ICH) with therapeutic anticoagulation complicates VTE treatment. Some retrospective studies have reported an increased risk of ICH associated with therapeutic anticoagulation; however, effective alternatives to anticoagulation are lacking. The aim of our study is to assess the risk of ICH in HGG patients with VTE on low molecular weight heparin (LMWH). METHODS We performed a retrospective matched cohort study of HGG patients from 1/2005-8/2016. Blinded review of neuroimaging for ICH was performed. For analysis of the primary endpoint, estimates of cumulative incidence (CI) of ICH were calculated using competing risk analysis with death as competing risk; significance testing was performed using the Gray's test. Median survival was estimated using Kaplan-Meier method. RESULTS 220 patients were included, 88 (40%) with VTE treated with LMWH, 22 (10%) with VTE, not on anticoagulation (AC), and 110 (50%) without VTE. A total of 43 measurable ICH was recorded: 19 (26%) in LMWH, 3 (14%) in VTE not on AC, and 21 (19%) in non-VTE cohort. No significant difference was observed in the 1-year CI of ICH in the LMWH cohort and non-AC with VTE group (17% versus 9%; Gray's test, p=0.36). Among patients without VTE, the 1-year CI of ICH was 13%. Median survival was similar among all three cohorts. CONCLUSIONS Our data suggest that therapeutic LMWH is not associated with substantially increased risk of ICH in HGG patients.
Collapse
Affiliation(s)
- Jasmin Jo
- Department of Internal Medicine, Division of Hematology and Oncology, East Carolina University/Vidant Medical Center, Greenville, NC
| | - Joseph Donahue
- Department of Radiology, University of Virginia, Charlottesville, VA
| | - Guneet Sarai
- Department of Primary Care and Population Health, Virginia Commonwealth University, Richmond, VA
| | - Gina Petroni
- Department of Public Health Sciences, Division of Biostatistics, University of Virginia, Charlottesville, VA
| | - David Schiff
- Department of Neurology, Division of Neuro-Oncology, University of Virginia, Charlottesville, VA
| |
Collapse
|
20
|
Saito M, Wages NA, Schiff D. Incidence, risk factors and management of venous thromboembolism in patients with primary CNS lymphoma. J Neurooncol 2021; 154:41-47. [PMID: 34164780 DOI: 10.1007/s11060-021-03791-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Venous thromboembolism (VTE) is a known complication of malignancy. While brain tumors in general predispose to VTE, the incidence in primary central nervous system lymphoma (PCNSL) is poorly characterized. We sought to characterize incidence, risk factors, management, and outcome of VTE in PCNSL METHOD: Retrospective study of 78 PCNSL patients from 2/1/2002 to 4/1/2020 at the University of Virginia RESULTS: 31% (24/78) of patients developed VTE. 12.8% (10/78) had deep venous thrombosis (DVT) alone, 11.5% (9/78) isolated pulmonary embolism (PE) and 6.4% (5/78) both. The median time from PCNSL diagnosis to VTE was 3 months. In a univariate competing risks analysis, previous VTE (p < 0.001), impaired ambulation (p = 0.035), baseline hemoglobin < 10 g/dL (p = 0.025) and history of diabetes mellitus (type 1 or 2) (p = 0.007) were associated with increased VTE risk. 34.8% were anticoagulated acutely with heparin (8/23) or 65.2% LMWH (15/23), and 25.0% (6/24) received warfarin, 41.7% (10/24) LMWH, and 33.3% (8/24) DOACs long-term. One adverse event was attributable to anticoagulation (arm hematoma with hemoglobin decrease). Five patients received IVC filters with concomitant oral anticoagulation; one experienced IVC thrombosis after anticoagulation discontinuation. Six of the 24 patients experienced recurrent VTE, four while anticoagulated. CONCLUSION Patients with PCNSL are at high risk of VTE, most of which accrues in the first few months. History of VTE, diabetes mellitus (type 1 or 2), impaired ambulatory status, or hemoglobin < 10 g/dL may predispose patients to this complication. While optimal management is uncertain, anticoagulation prevented recurrent VTE in most patients without intracranial bleeding.
Collapse
Affiliation(s)
- Miyabi Saito
- UVA School of Medicine, University of Virginia, Charlottesville, VA, 22903, USA
| | - Nolan A Wages
- Division of Translational Research & Applied Statistics, Department of Public Health Sciences, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22903-0717, USA
| | - David Schiff
- Divison of Neuro-Oncology, Department of Neurology, University of Virginia, PO Box 800432, Charlottesville, VA, 22908-0432, USA.
| |
Collapse
|
21
|
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
|