1
|
Lanting V, Oskam M, Wilmink H, Kamphuisen PW, van Es N. The role of germline and somatic mutations in predicting cancer-associated thrombosis: a narrative review. Curr Opin Hematol 2025; 32:138-145. [PMID: 39851266 PMCID: PMC11957438 DOI: 10.1097/moh.0000000000000861] [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] [Indexed: 01/26/2025]
Abstract
PURPOSE OF REVIEW Patients with cancer have an increased risk of venous thromboembolism (VTE). Guidelines suggest to use risk assessment tools to guide decisions about thromboprophylaxis, but current tools have modest discriminatory ability. Genetic information from the germline or tumor has the potential to improve VTE prediction. Here, we provide a clinical overview of the current role of genetics in cancer-associated VTE. RECENT FINDINGS Germline mutations, such as factor V Leiden and prothrombin G20210A, are associated with a 2- to 2.5-fold increased VTE risk in patients with cancer. Tumor-specific somatic mutations also contribute to VTE risk, such as ALK rearrangements increasing the risk in nonsmall cell lung cancer and IDH1 mutations decreasing the risk in gliomas. Other somatic mutations associated with VTE independent of tumor type include KRAS , STK11 , MET , KEAP1 , CTNNB1 , and CDKN2B . Incorporating data on germline or somatic mutations in risk scores improves discriminatory ability compared with the Khorana score. SUMMARY Specific germline and somatic mutations are associated with an increased VTE risk in patients with cancer and potentially improve performance of clinical risk scores. The increasing and widespread use of genetic testing in cancer care provides an opportunity for further development of prediction models incorporating genetic predictors.
Collapse
Affiliation(s)
- Vincent Lanting
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Merel Oskam
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| | - Hanneke Wilmink
- Amsterdam UMC, University of Amsterdam, department of Oncology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Pieter W. Kamphuisen
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
- Tergooi MC, department of Internal Medicine, Hilversum
| | - Nick van Es
- Amsterdam UMC, University of Amsterdam, department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam
| |
Collapse
|
2
|
Hao L, Zhang J, Di Y, Qi Z, Zhang P. Predicting a failure of postoperative thromboprophylaxis in non-small cell lung cancer: A stacking machine learning approach. PLoS One 2025; 20:e0320674. [PMID: 40168285 PMCID: PMC11960935 DOI: 10.1371/journal.pone.0320674] [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: 03/10/2024] [Accepted: 02/21/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) and its surgery significantly increase the venous thromboembolism (VTE) risk. This study explored the VTE risk factors and established a machine-learning model to predict a failure of postoperative thromboprophylaxis. METHODS This retrospective study included patients with NSCLC who underwent surgery between January 2018 and November 2022. The patients were randomized 7:3 to the training and test sets. Nine machine learning models were constructed. The three most predictive machine-learning classifiers were chosen as the first layer of the stacking machine-learning model, and logistic regression was the second layer of the meta-learning model. RESULTS This study included 362 patients, including 58 (16.0%) with VTE. Based on the multivariable logistic regression analysis, age, platelets, D-dimers, albumin, smoking history, and epidermal growth factor receptor (EGFR) exon 21 mutation were used to develop the nine machine-learning models. LGBM Classifier, RandomForest Classifier, and GNB were chosen for the first layer of the stacking machine learning model. The area under the received operating characteristics curve (ROC-AUC), accuracy, sensitivity, and specificity of the stacking machine learning model in the training/test set were 0.984/0.979, 0.949/0.954, 0.935/1.000, and 0.958/0.887, respectively. In the validation set, the final stacking machine learning model demonstrated an ROC AUC of 0.983, accuracy of 0.937, sensitivity of 0.978, and specificity of 0.947. The decision curve analyses revealed high benefits. CONCLUSION The stacking machine learning model based on EGFR mutation and clinical characteristics had a predictive value for postoperative VTE in patients with NSCLC.
Collapse
Affiliation(s)
- Ligang Hao
- Department of Thoracic Surgery, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Junjie Zhang
- Department of Computed Tomography and Magnetic Resonance, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Yonghui Di
- Department of Thoracic Surgery, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Zheng Qi
- Department of Clinical Lab, Xingtai People’s Hospital, Xingtai, Hebei, China
| | - Peng Zhang
- Department of Thoracic Surgery, Xingtai People’s Hospital, Xingtai, Hebei, China
| |
Collapse
|
3
|
Altwayan R, Tombuloglu H, Alhamid G, Karagoz A, Alshammari T, Alsaeed M, Al-Hariri M, Rabaan A, Unver T. Comprehensive review of thrombophilia: pathophysiology, prevalence, risk factors, and molecular diagnosis. Transfus Clin Biol 2025:S1246-7820(25)00047-3. [PMID: 40157494 DOI: 10.1016/j.tracli.2025.03.004] [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: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Thrombophilia, characterized by an imbalance between fibrinolysis and coagulation leading to inappropriate blood clotting, is a significant medical condition. The CDC has designated it as an underdiagnosed, serious, and potentially preventable disorder, contributing to an estimated 600,000-900,000 cases and 100,000 deaths annually in the United States. These figures surpass the combined annual mortality of AIDS, breast cancer, and motor vehicle accidents. The pathogenesis of thrombophilia involves complex interactions between genetic predispositions, such as mutations in Factor V Leiden, Factor II, MTHFR, and Serpine-1, and environmental factors, including unhealthy lifestyles, prolonged hospitalization, obesity, and cancer. Prevalence of specific genetic mutations varies across populations. Additional risk factors include age, family history, and pregnancy, with recent attention to increased susceptibility in SARS-CoV-2 infection. While molecular diagnostic techniques are available, there remains a need for robust, cost-effective, and accurate screening methods for large populations. This systematic review provides an updated overview of thrombophilia, encompassing pathophysiology, epidemiology, genetic and environmental risk factors, coagulation cascade, population-specific mutation prevalence, and diagnostic approaches. By synthesizing clinical and molecular evidence, this review aims to guide researchers, hematologists, and clinicians in the diagnosis and management of thrombophilia.
Collapse
Affiliation(s)
- Reham Altwayan
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia; Master Program of Biotechnology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Huseyin Tombuloglu
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia.
| | - Galyah Alhamid
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia
| | - Aysel Karagoz
- Quality Assurance Department, Turk Pharmaceutical and Serum Ind. Inc., Ankara, Turkey
| | - Thamer Alshammari
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia
| | - Moneerah Alsaeed
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia
| | - Mohammed Al-Hariri
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, 31441 Dammam, Saudi Arabia
| | - Ali Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Turgay Unver
- Faculty of Engineering, Ostim Technical University, Ankara 06374, Turkey
| |
Collapse
|
4
|
Furuya N, Tsubata Y, Hotta T, Yokoyama T, Yamasaki M, Ishikawa N, Fujitaka K, Kubota T, Kobayashi K, Isobe T. Arterial Thromboembolism in Patients With Advanced Lung Cancer: Secondary Analyses of the Rising-VTE/NEJ037 Study. Cancer Med 2025; 14:e70568. [PMID: 39783855 PMCID: PMC11714229 DOI: 10.1002/cam4.70568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 11/20/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Cancer-associated thromboembolism has been thoroughly investigated in previous studies, and direct oral anticoagulants (DOACs) were established for the treatment and prevention of venous thromboembolism (VTE). However, the risks of cancer-associated arterial thromboembolism (ATE) and the efficacy of DOACs remain unclear. OBJECTIVES To evaluate the risk factors and the clinical activity of edoxaban (EDO) for the prevention of ATE in patients with advanced lung cancer. METHODS From the prospective Rising-VTE/NEJ037 study which investigated VTE in newly diagnosed advanced lung cancer, we investigated the incidence rate and the risk factors of ATE as secondary endpoints. RESULTS A total of 1008 patients were screened for VTE at study baseline and were followed up for 2 years. Excluding patients with a contraindication to DOACs, those with VTE were treated with EDO. ATE events were identified in 41 patients (4.1%). The most common location for ATE was cerebral infarction (N = 31, 75.6%), followed by myocardial infarction (N = 4, 9.8%). Multivariate analysis determined the incidence of VTE, D-dimer, a comorbidity of atrial fibrillation, and four other factors as independent risk factors of ATE. For VTE (+) patients, the incidence rate of ATE was 15.9% for the EDO administration (+) patients, compared with 11.1% for the EDO administration (-) patients (p = 0.626). CONCLUSIONS The incidence rate of ATE was 4.1% over 2-year follow-up in advanced lung cancer patients. VTE was further identified as an independent risk factor for ATE, while intervention with DOACs was seen as less effective for the prevention of ATE in advanced lung cancer patients with VTE. TRIAL REGISTRATION This trial was registered in the Japan Registry of Clinical Trials (jRCTs061180025).
Collapse
Affiliation(s)
- Naoki Furuya
- Division of Respiratory Medicine, Department of Internal MedicineSt. Marianna University School of MedicineKawasakiJapan
| | - Yukari Tsubata
- Division of Medical Oncology and Respiratory Medicine, Department of Internal MedicineShimane University Faculty of MedicineIzumoJapan
| | - Takamasa Hotta
- Division of Medical Oncology and Respiratory Medicine, Department of Internal MedicineShimane University Faculty of MedicineIzumoJapan
| | - Toshihide Yokoyama
- Department of Respiratory MedicineKurashiki Central HospitalKurashikiJapan
| | - Masahiro Yamasaki
- Department of Respiratory DiseaseHiroshima Red Cross Hospital and Atomic‐Bomb Survivors HospitalHiroshimaJapan
| | - Nobuhisa Ishikawa
- Department of Respiratory MedicineHiroshima Prefectural HospitalHiroshimaJapan
| | - Kazunori Fujitaka
- Department of Respiratory MedicineHiroshima University HospitalHiroshimaJapan
| | - Tetsuya Kubota
- Department of Respiratory Medicine and AllergologyKochi University HospitalKochiJapan
| | - Kunihiko Kobayashi
- Department of Respiratory MedicineSaitama Medical University International Medical CenterSaitamaJapan
| | - Takeshi Isobe
- Division of Medical Oncology and Respiratory Medicine, Department of Internal MedicineShimane University Faculty of MedicineIzumoJapan
| |
Collapse
|
5
|
Yoshimine K, Tobino K, Obata Y, Sogabe S, Uchida K, Murakami Y, Yamamoto R. Trousseau's Syndrome in Lung Cancer Patients: A Retrospective Study in a Japanese Community Hospital. Cureus 2024; 16:e68400. [PMID: 39355485 PMCID: PMC11444742 DOI: 10.7759/cureus.68400] [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] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Trousseau's syndrome is a cancer-associated thromboembolism that significantly impacts patients' prognosis and quality of life (QOL). This study aimed to investigate the frequency, characteristics, and prognosis of Trousseau's syndrome in lung cancer patients at a Japanese community hospital and examine the effects of therapeutic agents on this condition. We retrospectively reviewed the electronic medical records of lung cancer patients diagnosed with thrombotic complications at the time of diagnosis in our department between August 2013 and April 2019. Patients' characteristics, thromboembolism sites, treatments, and prognosis were analyzed. Among 956 lung cancer patients, 19 (2%) had Trousseau's syndrome. The median age was 65 years, and adenocarcinoma was the most common histologic type (78.9%). The most common site of thromboembolism was the brain (84.2%). The median survival time was 84 days, and 52.6% of patients died within 90 days of diagnosis. Patients who survived longer than 90 days tended to have a higher frequency of non-adenocarcinoma histology, EGFR gene mutations, and therapeutic induction with immune checkpoint inhibitors (ICI). Trousseau's syndrome in lung cancer patients is associated with poor prognosis. Histologic type, EGFR mutation status, and treatment with ICI may influence the prognosis. Future larger-scale studies are needed to validate these potential prognostic factors and to develop personalized treatment strategies.
Collapse
Affiliation(s)
| | | | - Yumi Obata
- Respiratory Medicine, Iizuka Hospital, Fukuoka, JPN
| | | | | | | | | |
Collapse
|
6
|
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
|
7
|
Wojtukiewicz MZ, Tesarova P, Karetová D, Windyga J. In Search of the Perfect Thrombosis and Bleeding-Associated Cancer Scale. Semin Thromb Hemost 2024; 50:443-454. [PMID: 37852295 DOI: 10.1055/s-0043-1776003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Thrombosis and bleeding are commonly observed in cancer patients, and their management is crucial for positive patient outcomes. A comprehensive, prophylactic, and therapeutic management of venous thrombosis should focus on identifying the patients who would benefit most from treatment to reduce mortality and minimize the risk of thrombosis recurrence without significantly increasing the risk of bleeding. Existing cancer scales provide valuable information for assessing the overall burden of cancer and guiding treatment decisions, but their ability to predict thrombotic and bleeding events remains limited. With increasing knowledge of the pathophysiology of cancer and the availability of advanced anticancer therapies, new risk factors for cancer-associated thrombosis and bleeding are being identified. In this report, we analyze the current literature and identify new risk factors for venous thrombosis and bleeding which are not included in routinely used risk scores. While some existing cancer scales partially capture the risk of thrombosis and bleeding, there is a need for more specific and accurate scales tailored to these complications. The development of such scales could improve risk stratification, aid in treatment selection, and enhance patient care. Therefore, further research and development of novel cancer scales focused on thrombosis and bleeding are warranted to optimize patient management and outcomes.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, Bialystok, Poland
- Comprehensive Cancer Center, Bialystok, Poland
| | - Petra Tesarova
- Department of Oncology, Institute of Radiation Oncology, First Faculty of Medicine, Charles University and Bulovka University Hospital, Prague, Czech Republic
| | - Debora Karetová
- Second Department of Medicine-Department of Cardiovascular Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jerzy Windyga
- Department of Hemostasis Disorders and Internal Medicine, Laboratory of Hemostasis and Metabolic Diseases, Institute of Hematology and Transfusion Medicine in Warsaw, Warsaw, Poland
| |
Collapse
|
8
|
Charpidou A, Gerotziafas G, Popat S, Araujo A, Scherpereel A, Kopp HG, Bironzo P, Massard G, Jiménez D, Falanga A, Kollias A, Syrigos K. Lung Cancer Related Thrombosis (LCART): Focus on Immune Checkpoint Blockade. Cancers (Basel) 2024; 16:450. [PMID: 38275891 PMCID: PMC10814098 DOI: 10.3390/cancers16020450] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/10/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Cancer-associated thrombosis (CAT) is a common complication in lung cancer patients. Lung cancer confers an increased risk of thrombosis compared to other solid malignancies across all stages of the disease. Newer treatment agents, including checkpoint immunotherapy and targeted agents, may further increase the risk of CAT. Different risk-assessment models, such as the Khorana Risk Score, and newer approaches that incorporate genetic risk factors have been used in lung cancer patients to evaluate the risk of thrombosis. The management of CAT is based on the results of large prospective trials, which show similar benefits to low-molecular-weight heparins (LMWHs) and direct oral anticoagulants (DOACs) in ambulatory patients. The anticoagulation agent and duration of therapy should be personalized according to lung cancer stage and histology, the presence of driver mutations and use of antineoplastic therapy, including recent curative lung surgery, chemotherapy or immunotherapy. Treatment options should be evaluated in the context of the COVID-19 pandemic, which has been shown to impact the thrombotic risk in cancer patients. This review focuses on the epidemiology, pathophysiology, risk factors, novel predictive scores and management of CAT in patients with active lung cancer, with a focus on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Andriani Charpidou
- Third Department of Internal Medicine and Laboratory, Athens Medical School, National and Kapodistrian University of Athens, Sotiria General Hospital, 157 72 Athens, Greece; (A.K.)
| | - Grigorios Gerotziafas
- Assistance Publique-Hôpitaux de Paris, Thrombosis Center, Service D’Hématologie Biologique Hôpital Tenon, Sorbonne Université, 75005 Paris, France
| | - Sanjay Popat
- Royal Marsden Hospital, Institute of Cancer Research, London SW3 6JJ, UK
| | - Antonio Araujo
- Department of Medical Oncology, Centro Hospitalar Universitário do Porto, 4099-001 Porto, Portugal;
| | - Arnaud Scherpereel
- Department of Pulmonary and Thoracic Oncology, University of Lille, University Hospital (CHU), 59000 Lille, France;
| | - Hans-Georg Kopp
- Departments of Molecular Oncology and Thoracic Oncology, Robert-Bosch-Hospital Stuttgart, 70376 Stuttgart, Germany
| | - Paolo Bironzo
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, 10124 Turin, Italy
| | - Gilbert Massard
- Department of Thoracic Surgery, Hôpitaux Robert Schuman, 2540 Luxembourg, Luxembourg
| | - David Jiménez
- Respiratory Department, Ramón y Cajal Hospital, Instituto Ramón y Cajal de Investigación Sanitaria IRYCIS, 28034 Madrid, Spain;
| | - Anna Falanga
- Department of Transfusion Medicine and Hematology, Hospital Papa Giovanni XXIII, University of Milan Bicocca, 24129 Bergamo, Italy
| | - Anastasios Kollias
- Third Department of Internal Medicine and Laboratory, Athens Medical School, National and Kapodistrian University of Athens, Sotiria General Hospital, 157 72 Athens, Greece; (A.K.)
| | - Konstantinos Syrigos
- Third Department of Internal Medicine and Laboratory, Athens Medical School, National and Kapodistrian University of Athens, Sotiria General Hospital, 157 72 Athens, Greece; (A.K.)
| |
Collapse
|
9
|
Aguiar De Azevedo L, Orione C, Tromeur C, Couturaud F, Descourt R, Geier M. Incidence of venous thromboembolism and association with PD-L1 expression in advanced non-small cell lung cancer patients treated with first-line chemo-immunotherapy. Front Oncol 2024; 13:1221106. [PMID: 38260845 PMCID: PMC10800377 DOI: 10.3389/fonc.2023.1221106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Background Venous thromboembolism (VTE) is a serious complication in non-small cell lung cancer (NSCLC) patients. The use of thromboprophylactic therapy is subject to an accurate assessment of the VTE risk depending on patients, tumor characteristics and type of systemic antineoplastic treatments. However, little is known concerning the risk of VTE in patients suffering from an advanced NSCLC treated with first-line chemo-immunotherapy and the impact of tumor biomarkers such as PD-L1 expression. Methods We performed a retrospective, observational, single-centre study in a cohort of advanced NSCLC patients treated with first-line chemo-immunotherapy. The primary endpoint was the incidence of VTE. Secondary endpoints were the cumulative incidence of VTE, the impact of PD-L1 on VTE occurrence, overall survival, the rate of VTE recurrence under anticoagulant treatment and the rate of bleeding complications. Results 109 patients were included, of whom 21 (19.3%) presented a VTE event during a median follow-up of 13 months. VTE incidence at 3, 6 and 12 months was 12.1%, 15.1% and 17.5% respectively. 61% were pulmonary embolisms, 9.5% were isolated deep vein thrombosis and 14.3% were central venous catheter-related thrombosis. Our study did not show a significant impact of PD-L1 on VTE occurrence. Overall survival at 6, 12 and 24 months was 81.9%, 74.4% and 70.3% respectively. Four patients developed a recurrent VTE under anticoagulation therapy 3 to 5 months after the first VTE event. One patient suffered from a major bleeding complication while under anticoagulation therapy, leading to death. Conclusion VTE is a common complication in advanced NSCLC patients treated with concomitant chemo-immunotherapy. In our study, 19.3% of patients developed a VTE during a median follow-up of 13 months. PD-L1 did not appear to be associated with VTE occurrence. We recorded high VTE recurrence rates despite anticoagulant treatment. Further investigations are needed to determine if high PD-L1 expression is associated with VTE.
Collapse
Affiliation(s)
| | - Charles Orione
- Service de Pneumologie, Centre Hospitalier de Cornouaille, Quimper, France
| | - Cécile Tromeur
- Service de Pneumologie, Centre Hospitalier Regional Universitaire de Brest, Brest, Brittany, France
| | - Francis Couturaud
- Service de Pneumologie, Centre Hospitalier Regional Universitaire de Brest, Brest, Brittany, France
| | - Renaud Descourt
- Service d’oncologie médicale, Centre Hospitalier Regional Universitaire de Brest, Brest, Brittany, France
| | - Margaux Geier
- Service d’oncologie médicale, Centre Hospitalier Regional Universitaire de Brest, Brest, Brittany, France
| |
Collapse
|
10
|
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
|
11
|
Nwagha T, Nweke M. Stratification of Risk Factors of Lung Cancer-Associated Venous Thromboembolism and Determining the Critical Point for Preemptive Intervention: A Systematic Review With Meta-analysis. Clin Med Insights Oncol 2023; 17:11795549231175221. [PMID: 37426681 PMCID: PMC10328178 DOI: 10.1177/11795549231175221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/20/2023] [Indexed: 07/11/2023] Open
Abstract
Background Several biomarkers or risk factors have been identified and several prediction models exist. The major limitations inherent in these models include cost-ineffectiveness and lack of systematic stratification of risk factors resulting in the inclusion of clinically insignificant biomarkers in the models. This review aimed to systematically stratify the risk factors of lung cancer-associated venous thromboembolism (VTE) and determine the critical point for preemptive intervention. Methods This systematic review was structured as per the Preferred Reporting Item for Systematic Reviews and Meta-analyses. We searched MEDLINE, PubMed, Cochrane Library, CINAHL, Academic Search Complete, and PsycINFO from the onset to June 2022. We included studies that reported the risk factors of lung cancer-associated VTE and corresponding risk estimates, irrespective of treatment status but studies were excluded if patients were on anti-VTE medications. We employed random effects models of meta-analysis and computed risk stability index and risk weight (Rw) to achieve the review objectives. The review protocol is registered with PROSPERO (CRD42022336476). Results The clinically significant risk factors of VTE in lung cancer patients were D-dimer (odds ratio [OR] = 5.510, 95% CI = 2.6-11.7; Rw = 5.0), albumin (OR = 2.2, 95% CI = 1.0-4.8; Rw = 1.79), leukocyte (OR = 2.48, 95% CI = 1.9-3.2; Rw = 1.77), histological type (OR = 1.69 , 95% CI = 1.2-2.4; Rw = 1.3), age (OR = 1.56; Rw = 0.99), and hemoglobin (OR = 1.85, 95% CI = 1.3-2.6; Rw = 0.92). Based on the distribution of Rw across risk factors, the critical point (upper third of the upper quartile class) was 4.5 and may mark the point at which preemptive intervention should be commenced. Conclusions Targeted screening for VTE in lung cancer patients could be patient-specific and should be based on a combination of the most significant risk factors required to meet the critical point, provided that such a combination is affordable as illustrated in the ALBAH model. Registration The review protocol is registered with PROSPERO (ID: CRD42022336476).
Collapse
Affiliation(s)
- Theresa Nwagha
- Department of Haematology and
Immunology, Faculty of Medicine, University of Nigeria Teaching Hospital
Ituku-Ozalla, Enugu, Nigeria
| | - Martins Nweke
- Department of Physiotherapy, Evangel
University, Akaeze, Nigeria
- Fledgelight Evidence Consult, Enugu,
Nigeria
| |
Collapse
|
12
|
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: 3] [Impact Index Per Article: 1.5] [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
|
13
|
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
|
14
|
Feldman S, Gupta D, Navi BB, Grace Ho KW, Willeit P, Devlin S, Bolton KL, Arcila ME, Mantha S. Tumor Genomic Profile Is Associated With Arterial Thromboembolism Risk in Patients With Solid Cancer. JACC CardioOncol 2023; 5:246-255. [PMID: 37144118 PMCID: PMC10152200 DOI: 10.1016/j.jaccao.2023.01.009] [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: 06/24/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 05/06/2023] Open
Abstract
Background Patients with cancer have an increased risk for arterial thromboembolism (ATE). Scant data exist about the impact of cancer-specific genomic alterations on the risk for ATE. Objectives The aim of this study was to determine whether individual solid tumor somatic genomic alterations influence the incidence of ATE. Methods A retrospective cohort study was conducted using tumor genetic alteration data from adults with solid cancers who underwent Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets testing between 2014 and 2016. The primary outcome, ATE, was defined as myocardial infarction, coronary revascularization, ischemic stroke, peripheral arterial occlusion, or limb revascularization and identified through systematic electronic medical record assessments. Patients were followed from date of tissue-matched blood control accession to first ATE event or death, for up to 1 year. Cause-specific Cox proportional hazards regression was used to determine HRs of ATE for individual genes adjusted for pertinent clinical covariates. Results Among 11,871 eligible patients, 74% had metastatic disease, and there were 160 ATE events. A significantly increased risk for ATE independent of tumor type was noted for the KRAS oncogene (HR: 1.98; 95% CI: 1.34-2.94; multiplicity-adjusted P = 0.015) and the STK11 tumor suppressor gene (HR: 2.51; 95% CI: 1.44-4.38; multiplicity-adjusted P = 0.015). Conclusions In a large genomic tumor-profiling registry of patients with solid cancers, alterations in KRAS and STK11 were associated with an increased risk for ATE independent of cancer type. Further investigation is needed to elucidate the mechanism by which these mutations contribute to ATE in this high-risk population.
Collapse
Affiliation(s)
- Stephanie Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dipti Gupta
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Babak B. Navi
- Department of Neurology and the Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ka-Wai Grace Ho
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Peter Willeit
- Clinical Epidemiology Team, Medical University of Innsbruck, Innsbruck, Austria
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Sean Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kelly L. Bolton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria E. Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Simon Mantha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Address for correspondence: Dr Simon Mantha, Memorial Sloan Kettering Cancer Center, Koch Center, 530 East 74th Street, New York, New York 10021, USA.
| |
Collapse
|
15
|
Could PD-L1 positivity be associated with venous thrombosis in patients with non-small cell lung cancer? J Thromb Thrombolysis 2023; 55:382-391. [PMID: 36564589 DOI: 10.1007/s11239-022-02753-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2022] [Indexed: 12/25/2022]
Abstract
The risk of venous thromboembolism (VTE) is increased in non-small cell lung cancer (NSCLC), and defining at-risk patients is important. Thus, we aimed to assess the association between programmed cell death ligand 1 (PD-L1) expression and VTE [pulmonary embolism (PE), deep venous thrombosis (DVT)] in NSCLC. In this retrospective, observational multicentre study, 369 patients with NSCLC who had PD-L1 immunohistochemistry based on biopsies taken between January 2017 and December 2019, were divided as PD-L1-positive (n = 181) and -negative (n = 188) groups, and low-positive (n = 99) and high-positive (n = 82) PD-L1 groups. Among all population, 12.5% of them developed a VTE during a median follow-up of 474 days. The rates of DVT, PE, and PE + DVT were 5.7%, 6% and 0.8%, respectively. VTE (15.5% vs. 9.5%) and DVT (3.8% vs. 7.4%) were similar between two groups, while PE was significantly higher in PD‑L1-positive group than those in PD-L1-negative group (11.1% vs 1%, p < 0.001). There were no significant differences between low- and high-positive groups in terms of VTE (14.1% vs. 17%), PE (12.1% vs. 9.8%), and DVT (2% vs. 6.1%). In the multivariate analysis, multiple metastases (Hazard ratio [HR] 4.02; 95% confidence interval [Cl] 1.18-13.63; p = 0.07) and PD-L1 positivity was associated with an increased PE risk (HR 8.39; 95% Cl 2.07-34.07; p = 0.003). In conclusion, PD-L1 positivity may be of important role in predicting the increased risk of PE in patients with NSCLC and thereby may be used to define patients likely to benefit from thromboprophylaxis.
Collapse
|
16
|
Qin L, Liang Z, Xie J, Li X. Estimating Venous Thromboembolism Risk in Metastatic Colorectal Cancer Inpatients: Validation of Existing Risk Scores and Development of New Risk Scores. Clin Appl Thromb Hemost 2023; 29:10760296231196859. [PMID: 37691565 PMCID: PMC10498692 DOI: 10.1177/10760296231196859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Metastatic colorectal cancer (mCRC) patients are predisposed to venous thromboembolism (VTE). This study aimed to (1) evaluate the efficacy of 4 existing cancer-specific VTE models in predicting VTE incidence among hospitalized mCRC patients, and (2) examine the influence of incorporating mCRC molecular subtypes into these models. We conducted an evaluation of 4 cancer-specific VTE models, including Khorana, Vienna CATS, Protecht, and CONKO in a dataset involving 1392 mCRC patients. To evaluate the predictive performance, we utilized receiver operating characteristic (ROC) curves for both the original models and the modified models that incorporated microsatellite instability status or KRAS/NRAS/BRAF mutations. Moreover, we computed the net reclassification improvement (NRI) to quantify the enhancements made to the modified VTE risk models. All models demonstrated a moderate area under the ROC curve (ROC-AUC) when predicting the occurrence of VTE: Khorana (0.550), Vienna CATS (0.671), Protecht (0.652), and CONKO (0.578). The incorporation of KRAS and BRAF mutations significantly improved the ROC-AUC of all 4 existing models (modified Khorana: 0.796, modified Vienna CATS: 0.832, modified Protecht: 0.834, and modified CONKO: 0.809). After dichotomizing the risk using a threshold of 3 points and comparing them with the original models, NRI values for the 4 modified models were 0.97, 0.95, 1.11, and 0.98, respectively. All 4 cancer-specific VTE models exhibit moderate performance when identifying mCRC patients at high risk of VTE. Incorporating KRAS and BRAF mutations may enhance the prediction of VTE in hospitalized mCRC patients.
Collapse
Affiliation(s)
- Li Qin
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhikun Liang
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jingwen Xie
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoyan Li
- Department of Pharmacy, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Tawil N, Rak J. Blood coagulation and cancer genes. Best Pract Res Clin Haematol 2022; 35:101349. [DOI: 10.1016/j.beha.2022.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
|
18
|
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]
|
19
|
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: 2.7] [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
|
20
|
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
|
21
|
Athanazio RA, Ceresetto JM, Marfil Rivera LJ, Cesarman-Maus G, Galvez K, Marques MA, Tabares AH, Ortiz Santacruz CA, Santini FC, Corrales L, Cohen AT. Direct Oral Anticoagulants for the Treatment of Cancer-Associated Venous Thromboembolism: A Latin American Perspective. Clin Appl Thromb Hemost 2022; 28:10760296221082988. [PMID: 35261295 PMCID: PMC8918974 DOI: 10.1177/10760296221082988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Venous thromboembolism (VTE) is a leading cause of morbidity and mortality in patients with cancer. On the basis of results from randomized controlled trials, direct oral anticoagulants (DOACs) are now recommended for the treatment of cancer-associated VTE. The decision to use a DOAC requires consideration of bleeding risk, particularly in patients with gastrointestinal (GI) malignancies, the cost-benefit and convenience of oral therapy, and patient preference. While efficacy with apixaban, edoxaban, and rivaroxaban versus dalteparin has been consistent in the treatment of cancer-associated VTE, heterogeneity is evident with respect to major GI bleeding, with an increased risk with edoxaban and rivaroxaban but not apixaban. Although cost and accessibility vary in different countries of Latin America, DOACs should be considered for the long-term treatment of cancer-associated VTE in all patients who are likely to benefit. Apixaban may be the preferred DOAC in patients with GI malignancies and LMWH may be preferred for patients with upper or unresected lower GI tumors. Vitamin K antagonists should only be used for anticoagulation when DOACs and low molecular weight heparin are inaccessible or unsuitable.
Collapse
Affiliation(s)
- Rodrigo Abensur Athanazio
- Pulmonary Division-Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
- Rodrigo Abensur Athanazio, Pulmonary Division-Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina de São Paulo,
Av. Dr. Enéas de Carvalho Aguiar, 44 - 5 andar (pneumologia), 05403-900 - São Paulo/SP – Brazil.
| | | | - Luis Javier Marfil Rivera
- Servicio de Hematología, Hospital Universitario “Dr José E González”, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | | | - Kenny Galvez
- Cancer Unit, Hospital Pablo Tobón Uribe, Medellín, Colombia
| | - Marcos Arêas Marques
- Unit of Angiology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aldo Hugo Tabares
- Vascular Medicine and Thrombosis Service, Instituto Universitario de Ciencias Biomedicas de Córdoba (IUCBC), Córdoba, Argentina
| | | | | | - Luis Corrales
- Medical Oncology, Centro de Investigación y Manejo del Cáncer (CIMCA), San José, Costa Rica
| | - Alexander T. Cohen
- Department of Haematological Medicine, Guy's and St Thomas' Hospitals NHS Foundation Trust, King's College London, UK
| |
Collapse
|
22
|
Di W, Xu H, Xue T, Ling C. Advances in the Prediction and Risk Assessment of Lung Cancer-Associated Venous Thromboembolism. Cancer Manag Res 2021; 13:8317-8327. [PMID: 34764694 PMCID: PMC8575248 DOI: 10.2147/cmar.s328918] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022] Open
Abstract
According to the most recent data from the National Cancer Center, venous thromboembolism (VTE) has unsurprisingly become one of the most common complications in lung cancer. VTE not only interferes with the equilibrium of the clotting system but it also affects tumor progression and prognosis. For the identification of high-risk patients, many clinical risk assessment models have been developed and validated based on the risk factors found in previous studies. In this review, we will summarize advances in prediction and risk assessment of VTE, with a focus on early diagnosis and therapy, reduction of mortality, and the burden of medical costs in lung cancer patients.
Collapse
Affiliation(s)
- Wenjuan Di
- Department of Respiratory and Critical Care Medicine, The First Hospital Affiliated of Soochow University, Suzhou City, Jiangsu Province, People’s Republic of China
| | - Haotian Xu
- Department of Respiratory and Critical Care Medicine, The First Hospital Affiliated of Soochow University, Suzhou City, Jiangsu Province, People’s Republic of China
| | - Ting Xue
- Department of Respiratory and Critical Care Medicine, The First Hospital Affiliated of Soochow University, Suzhou City, Jiangsu Province, People’s Republic of China
| | - Chunhua Ling
- Department of Respiratory and Critical Care Medicine, The First Hospital Affiliated of Soochow University, Suzhou City, Jiangsu Province, People’s Republic of China
| |
Collapse
|
23
|
Choi JU, Lee NK, Seo H, Chung SW, Al-Hilal TA, Park SJ, Kweon S, Min N, Kim SK, Ahn S, Kim UI, Park JW, Kang CY, Kim IS, Kim SY, Kim K, Byun Y. Anticoagulation therapy promotes the tumor immune-microenvironment and potentiates the efficacy of immunotherapy by alleviating hypoxia. J Immunother Cancer 2021; 9:jitc-2021-002332. [PMID: 34341129 PMCID: PMC8330593 DOI: 10.1136/jitc-2021-002332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose Here, this study verifies that cancer-associated thrombosis (CAT) accelerates hypoxia, which is detrimental to the tumor immune microenvironment by limiting tumor perfusion. Therefore, we designed an oral anticoagulant therapy to improve the immunosuppressive tumor microenvironment and potentiate the efficacy of immunotherapy by alleviating tumor hypoxia. Experimental design A novel oral anticoagulant (STP3725) was developed to consistently prevent CAT formation. Tumor perfusion and hypoxia were analyzed with or without treating STP3725 in wild-type and P selectin knockout mice. Immunosuppressive cytokines and cells were analyzed to evaluate the alteration of the tumor microenvironment. Effector lymphocyte infiltration in tumor tissue was assessed by congenic CD45.1 mouse lymphocyte transfer model with or without anticoagulant therapy. Finally, various tumor models including K-Ras mutant spontaneous cancer model were employed to validate the role of the anticoagulation therapy in enhancing the efficacy of immunotherapy. Results CAT was demonstrated to be one of the perfusion barriers, which fosters immunosuppressive microenvironment by accelerating tumor hypoxia. Consistent treatment of oral anticoagulation therapy was proved to promote tumor immunity by alleviating hypoxia. Furthermore, this resulted in decrease of both hypoxia-related immunosuppressive cytokines and myeloid-derived suppressor cells while improving the spatial distribution of effector lymphocytes and their activity. The anticancer efficacy of αPD-1 antibody was potentiated by co-treatment with STP3725, also confirmed in various tumor models including the K-Ras mutant mouse model, which is highly thrombotic. Conclusions Collectively, these findings establish a rationale for a new and translational combination strategy of oral anticoagulation therapy with immunotherapy, especially for treating highly thrombotic cancers. The combination therapy of anticoagulants with immunotherapies can lead to substantial improvements of current approaches in the clinic.
Collapse
Affiliation(s)
- Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, South Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Gwanak-gu, South Korea
| | - Na Kyeong Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Gwanak-gu, South Korea
| | - Hyungseok Seo
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Seung Woo Chung
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Taslim A Al-Hilal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas, USA
| | - Seong Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Gwanak-gu, South Korea
| | - Seho Kweon
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Nuri Min
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Sang Kyoon Kim
- Laboratory Animal Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Seohyun Ahn
- ST Pharm Research & Development Center, Siheung-si, South Korea
| | - Uk-Il Kim
- ST Pharm Research & Development Center, Siheung-si, South Korea
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam, South Korea
| | - Chang-Yuil Kang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Gwanak-gu, South Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seongbuk-gu, South Korea
| | - Sang Yoon Kim
- College of Medicine, University of Ulsan, Ulsan, South Korea
- Department of Otolaryngology, Asan Medical Center, Seoul, South Korea
| | - Kyungjin Kim
- ST Pharm Research & Development Center, Siheung-si, South Korea
| | - Youngro Byun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Gwanak-gu, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| |
Collapse
|
24
|
Papadopoulos V, Tsapakidis K, Markou A, Kokkalis A, Aidarinis C, Kotsakis A. New prophylaxis strategies to reduce the risk of thromboembolism in cancer. Expert Rev Anticancer Ther 2021; 21:1135-1144. [PMID: 34139938 DOI: 10.1080/14737140.2021.1941889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION : Patients with cancer are at risk of thrombotic events, mainly deep vein thrombosis and/or pulmonary embolism. The thrombosis risk is generally 4-6 times higher than in a healthy population and depends on factors related to patient characteristics, tumor factors, and treatment-related factors. The decision-making for prophylactic anticoagulation is individualized according to the relative risks and benefits. The VTE risk has been quantified using different assessment scores. In recent years, an effort has been made to establish "risk assessment models" specifically for patients undergoing chemotherapy. AREAS COVERED This article reviews current data and ongoing research on predictive factors involved in cancer-related thrombosis and it is highlighted the currently suggested strategies for prophylaxis. Several trials that compared the two treatment options, direct factor Xa inhibitor or LMWH, with placebo and not each other are discussed. In this article, was analyzed the safety and efficacy features that led several international organizations such as ASCO, NCCN, and others, to issue guidelines for the prophylaxis and treatment of patients at high risk of thrombosis by using LMWH, fondaparinux and DOACs. EXPERT OPINION ASCO, NCCN, and other international organizations recommend thromboprophylaxis in high risk patients. However, further investigation is needed to define better biomarkers for more accurate identification of cancer patients that will benefit from anticoagulant treatment.
Collapse
Affiliation(s)
- Vasileios Papadopoulos
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Konstantinos Tsapakidis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Alexandra Markou
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | - Alexandros Kokkalis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece
| | | | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larissa, Larissa, Thessaly, Greece.,Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Thessaly, Greece
| |
Collapse
|
25
|
Tawil N, Spinelli C, Bassawon R, Rak J. Genetic and epigenetic regulation of cancer coagulome - lessons from heterogeneity of cancer cell populations. Thromb Res 2021; 191 Suppl 1:S99-S105. [PMID: 32736787 DOI: 10.1016/s0049-3848(20)30405-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/05/2020] [Accepted: 01/12/2020] [Indexed: 12/15/2022]
Abstract
Cancer-associated thrombosis (CAT) is a morbid, potentially life threatening and biologically impactful paraneoplastic state. At least in part, CAT is likely driven by cancer-specific mechanisms the nature of which is still poorly understood, hampering diagnostic, prophylactic and therapeutic efforts. It is increasingly appreciated that cancer-specific drivers of CAT include a constellation of oncogenic mutations and their superimposed epigenetic states that shape the transcriptome, phenotype and secretome of cancer cell populations, including the repertoire of genes impacting the vascular and coagulation systems. High-grade brain tumours, such as glioblastoma multiforme (GBM) represent a paradigm of locally initiated haemostatic abnormalities that propagate systemically, likely through circulating mediators, such as extracellular vesicles and soluble factors. Reciprocally, CAT impacts the biology of cancer cells and may drive tumour evolution. The constituent, oncogene-transformed cancer cell populations form complex ecosystems, the intricate architecture of which has been recently revealed by single cell sequencing technologies. Amidst this phenotypic heterogeneity, several alternative pathways of CAT may exist both between and within individual tumours and their subtypes, including GBM. Indeed, different contributions of cells expressing key coagulant mediators, such as tissue factor, or podoplanin, have been identified in GBM subtypes driven by oncogenic mutations in EGFR, IDH1 and other transforming genes. Thus, a better understanding of cellular sources of CAT, including dominant cancer cell phenotypes and their dynamic shifts, may help design more personalised approaches to thrombosis in cancer patients to improve outcomes.
Collapse
Affiliation(s)
- Nadim Tawil
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Cristiana Spinelli
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Rayhaan Bassawon
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Janusz Rak
- McGill University, Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
26
|
Qian X, Fu M, Zheng J, Zhou J, Zhou J. Driver Genes Associated With the Incidence of Venous Thromboembolism in Patients With Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:680191. [PMID: 33996610 PMCID: PMC8117344 DOI: 10.3389/fonc.2021.680191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The association between driver genes and the incidence of thromboembolic events (TEs) in patients diagnosed with non-small-cell lung cancer (NSCLC) needs to be quantified to guide clinical management. METHODS We interrogated PubMed, Embase, Web of Science and Cochrane library databases for terms related to venous thromboembolism (VTE) and arterial thromboembolism (ATE) in patients diagnosed with non-small-cell lung cancer harboring driver genes. This search was conducted for studies published between 1 January, 2000 and 31 December, 2020. A random-effects meta-analysis was performed to analyze the pooled incidence and odds ratios of VTE in patients with different driver genes. RESULTS Of the 2,742 citations identified, a total of 25 studies that included 21,156 patients met eligibility criteria. The overall pooled incidence of VTE in patients with driver genes was 23% (95% CI 18-29). Patients with ROS1 rearrangements had the highest incidence of VTE (37%, 95%CI 23-52). ALK rearrangements were associated with increased VTE risks (OR=2.08,95% CI 1.69-2.55), with the second highest incidence of VTE (27%, 95%CI 20-35). Both groups of patients with EGFR and KRAS mutations did not show a significantly increased risk for VTE (OR=1.33, 95% CI 0.75-2.34; OR=1.31, 95% CI 0.40-4.28). CONCLUSIONS ALK rearrangements were shown to be associated with increased VTE risks in patients diagnosed with non-small lung cancer, while there was no significant relation observed between VTE risks and EGFR or KRAS mutations in lung cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
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: 13.3] [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
|
28
|
Liu Y, Wang W, Wu F, Gao G, Xu J, Li X, Zhao C, Yang S, Mao S, Pan Y, Jia K, Shao C, Chen B, Ren S, Zhou C. High discrepancy in thrombotic events in non-small cell lung cancer patients with different genomic alterations. Transl Lung Cancer Res 2021; 10:1512-1524. [PMID: 33889526 PMCID: PMC8044490 DOI: 10.21037/tlcr-20-1290] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Acute complications, such as venous thromboembolism (VTE), are common in patients with advanced severe lung cancers. However, current VTE risk scores cannot adequately identify high-risk patients with non-small cell lung cancer (NSCLC). The study proposed to elucidated the incidence of thromboembolism (TE) in patients with different oncogenic aberrations and the impact of these aberrations on the efficacy of targeted therapy in patients with NSCLC. Methods A systemic review was conducted in Web of Science, PubMed, Embase and the Cochrane Library to evaluate the incidence of TE in different molecular subtypes of NSCLC. Data from patients diagnosed of advanced NSCLC who harboring anaplastic lymphoma kinase (ALK) or ROS proto-oncogene 1 receptor tyrosine kinase (ROS1) rearrangements since 2016 to 2019 were also retrospectively collected. A meta-analysis with random-effects model, sensitivity analysis and publication bias were performed. The principal summary measure was incidence of thrombotic events in NSCLC patients. And the efficacy of tyrosine kinase inhibitor (TKI) therapy was compared between the two subgroups. Results A total of 5,767 cases from 20 studies were included in the analysis of the incidence of thrombosis in patients with different oncogenic alterations. The pooled analysis showed a higher risk of thrombosis in ROS1-fusion types (41%, 95% CI: 35-47%) and ALK-fusion types (30%, 95% CI: 24-37%) than in EGFR-mutation (12%, 95% CI: 8-17%), KRAS-mutation (25%, 95% CI: 13-50%), and wild-type (14%, 95% CI: 10-20%) cases. A high prevalence of thrombosis (ALK: 24.4%; ROS1: 32.6%) was observed in the Shanghai Pulmonary Hospital (SPH) cohort of 224 patients with ALK or ROS1 fusion. Furthermore, patients with embolism had significantly shorter progression-free survival (PFS) after TKI therapy than those without embolism, both in the ALK+ cohort (5.6 vs. 12.9 months, P<0.0001) and in the ROS1+ cohort (9.6 vs. 17.6 months, P=0.0481). Conclusions NSCLC patients with ALK/ROS1 rearrangements are more likely to develop thrombosis than patients with other oncogenic alterations. Thrombosis may also be associated with an inferior response and PFS after TKI therapy.
Collapse
Affiliation(s)
- Yiwei Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wanying Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shuo Yang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yingying Pan
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chuchu Shao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
29
|
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: 3.4] [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
|
30
|
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: 4.0] [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
|
31
|
Xiong W, Du H, Ding W, Sun J, Xu M, Guo X. The association between pulmonary embolism and the cancer-related genomic alterations in patients with NSCLC. Respir Res 2020; 21:185. [PMID: 32677947 PMCID: PMC7364644 DOI: 10.1186/s12931-020-01437-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
To date, the association between the acute pulmonary embolism (PE) and the currently existing cancer-related genomic alterations in patients with non-small cell lung cancer (NSCLC) has been understudied. We reviewed patients with a confirmed histopathological diagnosis of NSCLC who underwent computed tomography pulmonary angiography (CTPA) and molecular tests including ALK, ROS1, EGFR, BRAF V600E as well as PD-L1 during the diagnosis of NSCLC, to explore the association between the genomic alterations and PE. The results showed that, for the patients with positive results of genomic alterations, the proportion of positive ALK (13.6%vs8.5%, P<0.001) and PD-L1 (24.7%vs19.9%, P = 0.001) in PE group were more than those in Non-PE group. The patients with positive ALK and PD-L1 had the most (19.0%) and second most (15.4%) incidence of PE among all the patients being studied. A multivariate Logistic regression analysis showed that the positive ALK [1.685(1.065-2.215)(P<0.001)] and PD-L1[1.798(1.137-2.201)(P<0.001)] were correlated with the occurrence of PE. The positive results of ALK and PD-L1 genomic alterations may indicate an increased risk of pulmonary embolism in patients with NSCLC.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Shanghai, 200092, Yangpu District, China.
| | - He Du
- Department of Oncology, Shanghai Pulmonary Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Wei Ding
- Department of Pulmonary and Critical Care Medicine, Punan Hospital, Pudong New District, Shanghai, China
| | - Jinyuan Sun
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Shanghai, 200092, Yangpu District, China
| | - Mei Xu
- Department of General Practice, North Bund Community Health Center, Shanghai, Hongkou District, China.
| | - Xuejun Guo
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Shanghai, 200092, Yangpu District, China.
| |
Collapse
|
32
|
Ortega Morán L, García Alfonso P, Aguilar Caballero I, Morón García B, Tirado Anula V, de Toro Carmena M, Soto Alsar J, Gutiérrez Alonso N, Bringas Beranek M, Martín Jiménez M, Muñoz Martín AJ. Incidence of venous thromboembolism in patients with colorectal cancer according to oncogenic status. Clin Transl Oncol 2020; 22:2026-2031. [PMID: 32270416 DOI: 10.1007/s12094-020-02339-1] [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] [Received: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND There are conflicting data regarding the role of KRAS mutation on the risk of venous thromboembolism (VTE) in colorectal cancer (CRC) patients. Moreover, the role of other biomarkers such as NRAS or BRAF has not been studied. PURPOSE To analyze the incidence of VTE in a cohort of patients with CRC based on KRAS, NRAS, and BRAF status. METHODS We performed a retrospective review of patients with unresectable locally advanced and metastatic CRC (mCRC) and known KRAS/NRAS/BRAF status, attended in the Medical Oncology Department of the Hospital General Universitario Gregorio Marañón (Madrid, Spain). The primary outcome was VTE defined as any venous thromboembolic event that occurred either 6 months before or at any time after the diagnosis of CRC. The biomarker status (KRAS, NRAS, and BRAF) and other predictors of thrombosis were collected. RESULTS One hundred and ninety-four patients were identified and included in the analysis. Forty-one patients (21.1%) experienced VTE. The incidence was 19.1% in RAS-mutated patients, 28.6% in BRAF-mutated patients and 21% in triple wild-type patients (p = NS). In multivariate analysis, ECOG ≥ 2 was the only independent predictor of VTE (OR 8.73; CI 95% 1.32-57.82; p = 0.025). CONCLUSIONS In our study, biomarkers have not been associated with an increased risk of VTE in CRC patients. A high incidence of VTE in BRAF-mutated patients has been observed and should be explored in further studies.
Collapse
Affiliation(s)
- L Ortega Morán
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España.,Cancer and Thrombosis Section, Spanish Society of Medical Oncology (SEOM), Madrid, Spain
| | - P García Alfonso
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - I Aguilar Caballero
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - B Morón García
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - V Tirado Anula
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - M de Toro Carmena
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - J Soto Alsar
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - N Gutiérrez Alonso
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - M Bringas Beranek
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - M Martín Jiménez
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España
| | - A J Muñoz Martín
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, España. .,Cancer and Thrombosis Section, Spanish Society of Medical Oncology (SEOM), Madrid, Spain.
| |
Collapse
|
33
|
Wang J, Hu B, Li T, Miao J, Zhang W, Chen S, Sun Y, Cui S, Li H. The EGFR-rearranged adenocarcinoma is associated with a high rate of venous thromboembolism. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:724. [PMID: 32042740 PMCID: PMC6990022 DOI: 10.21037/atm.2019.12.24] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 11/15/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND The purpose of this study was to investigate the incidence of venous thromboembolism (VTE) in epidermal growth factor receptor (EGFR) mutations patients with lung adenocarcinoma, to provide clinical basis for the perioperative prevention and treatment of VTE in patients with lung cancer. METHODS This study included patients with invasive lung adenocarcinoma confirmed by pathology from July 2016 to March 2018 after surgical pulmonectomy in Thoracic Surgery Department of Beijing Chaoyang Hospital. All enrolled patients were tested for relevant gene mutations. All patients were classified as adenocarcinoma subtypes by the 2011 International Association for the Study of Lung Cancer (IASLC), American Thoracic Society (ATS) and European Respiratory Society (ERS). Patients were divided into the VTE group and the control group according to whether VTE occurred postoperatively. Baseline data, gene test results, operative data and tumor pathology data between the two groups were compared. RESULTS According to the inclusion criteria, a total of 323 patients underwent lung cancer surgery were analyzed in this study, including 148 males and 175 females, aged from 25 to 82 years old. Postoperative VTE occurred in 33 patients, with an incidence of 10.2%. Compared the baseline data, there were significant differences in age and BMI between the two groups, but no significant differences in other indicators. Comparing the results of postoperative genetic tests, the cases of exon 18, 19, 20, 21, 30, 31 mutation, exon 18 and 20 mixed mutation and exon 20 and 21 mixed mutation were 5, 42, 6, 57, 1, 3, 1 and 1. The total EGFR mutation rate in the enrolled patients was 36.2% (117/323). Among them, the proportion of EGFR mutation in the VTE group was significantly higher than that in the non-VTE group (60.6% vs. 33.4%, P=0.002). Exon mutations in specific regions and mixed region of EGFR were not statistically significant between them; there was no statistical difference in the concomitant KRAS and ALK gene mutations between them. Comparing the pathological conditions, the proportion of acinar dominant lung adenocarcinoma in the VTE group was higher than that in the non-VTE group (57.6% vs. 30.7%, P=0.002); other histologic subtypes showed no statistical difference. The D-dimer difference before and 1 day after surgery, preoperative FEV1, surgical method, duration of surgery and blood loss were statistically significant differences between the two groups. The results of univariate analysis showed that there were significant differences between the VTE group and the control group in proportion of EGFR mutant lung adenocarcinoma, age, BMI, D-dimer difference before and 1 day after surgery, preoperative FEV1, surgical method, duration of surgery, blood loss and proportion of acinar dominant lung adenocarcinoma (P<0.05). However, VTE was not significantly correlated with gender, ALK or KRAS gene mutation and other factors. Multi-factor logistics regression analysis shows that Patients with EGFR gene mutation infiltrating lung adenocarcinoma, acinar dominant lung adenocarcinoma, FEV1 and difference of D-dimer (d1-pre) are independent risk factors for postoperative lung cancer complicated with VTE. CONCLUSIONS The incidence of VTE was 10.2% in patients with invasive lung adenocarcinoma without prophylactic anticoagulant therapy. EGFR gene mutation is an independent risk factor for postoperative VTE in lung cancer, and the incidence of VTE in adenocarcinoma with alveolar predominance is the highest. Other independent risk factors included the difference of D-dimer (d1-pre) and preoperative FEV1.
Collapse
Affiliation(s)
- Jing Wang
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Tong Li
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jinbai Miao
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wenqian Zhang
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Shuo Chen
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yixin Sun
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Songping Cui
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hui Li
- Department of Thoracic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
34
|
ROS1-rearranged Non-small-cell Lung Cancer is Associated With a High Rate of Venous Thromboembolism: Analysis From a Phase II, Prospective, Multicenter, Two-arms Trial (METROS). Clin Lung Cancer 2019; 21:15-20. [PMID: 31607443 DOI: 10.1016/j.cllc.2019.06.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/09/2019] [Accepted: 06/07/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Patients with cancer are at increased risk for venous thromboembolism (VTE), and 8% to 15% of patients with advanced non-small-cell lung cancer (NSCLC) experience a VTE event during the course of their disease. The incidence of VTE in molecularly defined NSCLC subgroups is still unclear. In this study, we investigated the incidence and the clinical correlates of VTE in patients with ROS1-rearranged NSCLC enrolled in the METROS trial (NCT02499614). PATIENTS AND METHODS The METROS trial is a prospective phase II study designed to assess efficacy, safety, and tolerability of crizotinib in patients with pre-treated metastatic NSCLC ROS1 rearrangement (cohort A) or with MET amplification or MET exon 14 mutation (cohort B). Patients with ROS1-rearranged NSCLC enrolled within cohort A and the expansion cohort of the trial were included in the primary analysis. RESULTS Among 48 patients with ROS1-rearranged NSCLC enrolled in the METROS study, 20 (41.6%) of 48 had at least 1 VTE event. Among them, 7 (35%) of 20 patients had ≥ 2 VTE events. VTE events consisted of pulmonary embolism (46.4%), deep vein thrombosis (39.2%), renal vein thrombosis (7.1%), internal jugular thrombosis (3.5%), and peripheral inserted central catheter-related thrombosis (3.5%). VTE events occurred at disease progression in 35.7% of cases, at diagnosis in 32.1% of cases, and during chemotherapy or crizotinib in 17.8% and 14.2%, respectively. CONCLUSION The incidence of VTE is 3- to 5-fold higher in patients harboring ROS1-rearrangment than previously observed for the general population with NSCLC. Larger studies are warranted to confirm our findings and determine whether the molecular profile of NSCLC should be incorporated into a risk-stratification tool and decision-making algorithm for VTE diagnosis and prophylaxis.
Collapse
|
35
|
Debourdeau P, Simonin C, Carbasse C, Debourdeau T, Zammit C, Scotté F. [Primary prophylaxis of venous thromboembolism in ambulatory cancer patients treated with antineoplastic agents]. Rev Med Interne 2019; 40:523-532. [PMID: 30928244 DOI: 10.1016/j.revmed.2019.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 11/29/2022]
Abstract
Apart from myeloma, primary prophylaxis of venous thromboembolism (VTE) in ambulatory cancer patients treated with chemotherapy is underused, despite its proven benefit for pancreatic cancer and to a lesser extent for lung cancer. This prophylaxis has been showed to be effective for myeloma, pancreas but in absolute numbers these cancers lead to a few venous thromboembolic events. Up to date, VTE risk scores cannot be used as a discriminatory criterion to select a high-risk population that could really benefit from this prevention. VTE depends in part on oncogenic mutations of tumor cells that result in an imbalance between activation and inhibition pathways that are involved in venous thrombus formation. So, stratification of risk of VTE in cancer patients could be considered from a clinical and molecular point of view and result in a tailored prophylaxis. This "personalized medicine" that is currently used for the anti-tumor treatment of many cancers and hematological malignancies, could lead to a more effective prophylaxis of VTE in cancer patients.
Collapse
Affiliation(s)
- P Debourdeau
- Institut Sainte Catherine, 250, chemin de Baigne-Pieds, CS 80005, 84918 Avignon cedex 09, France.
| | - C Simonin
- Institut Sainte Catherine, 250, chemin de Baigne-Pieds, CS 80005, 84918 Avignon cedex 09, France
| | - C Carbasse
- Institut Sainte Catherine, 250, chemin de Baigne-Pieds, CS 80005, 84918 Avignon cedex 09, France
| | - T Debourdeau
- Faculté de médecine Lyon est, université Claude Bernard Lyon 1, 8, boulevard Rockefeller, 69008 Lyon, France
| | - C Zammit
- Hôpitaux des Portes de Camargue, route d'Arles, 13150 Tarascon, France
| | - F Scotté
- Hôpital Foch, 40, rue Worth, BP 36, 92151 Suresnes cedex, France
| |
Collapse
|
36
|
Fernandes CJ, Morinaga LTK, Alves JL, Castro M, Calderaro D, Jardim CV, Souza R. Cancer-associated thrombosis: the when, how and why. Eur Respir Rev 2019; 28:28/151/180119. [PMID: 30918022 PMCID: PMC9488553 DOI: 10.1183/16000617.0119-2018] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer-associated thrombosis (CAT) is a condition in which relevance has been increasingly recognised both for physicians that deal with venous thromboembolism (VTE) and for oncologists. It is currently estimated that the annual incidence of VTE in patients with cancer is 0.5% compared to 0.1% in the general population. Active cancer accounts for 20% of the overall incidence of VTE. Of note, VTE is the second most prevalent cause of death in cancer, second only to the progression of the disease, and cancer is the most prevalent cause of deaths in VTE patients. Nevertheless, CAT presents several peculiarities that distinguish it from other VTE, both in pathophysiology mechanisms, risk factors and especially in treatment, which need to be considered. CAT data will be reviewed in this review. Cancer-associated thrombosis (CAT) presents peculiar features (risk factors and pathophysiology) that distinguish it from common VTE cases. Treatment of CAT requires a different approach, since the patients are more prone to recurrence and bleeding.http://ow.ly/j1Lu30nYmd5
Collapse
|
37
|
Tawil N, Chennakrishnaiah S, Bassawon R, Johnson R, D'Asti E, Rak J. Single cell coagulomes as constituents of the oncogene-driven coagulant phenotype in brain tumours. Thromb Res 2018; 164 Suppl 1:S136-S142. [PMID: 29703472 DOI: 10.1016/j.thromres.2018.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
Molecular profiling of human cancers revealed a startling diversity in disease-causing mechanisms superseding histological and anatomical commonalities. The emerging molecular subtypes and disease entities are often driven by distinct oncogenic pathways and their effectors, including those acting extracellularly on the vascular and coagulation systems. Indeed, several oncogenic mutations such as those affecting protein-coding genes (RAS, EGFR, PTEN, TP53) and non-coding RNA (microRNA) regulate multiple effectors of the coagulation system (coagulome), including tissue factor, protease activated receptors, clotting factors, mediators of platelet function and fibrinolysis. This is exemplified by differential coagulome profiles in the molecular subtypes of glioblastoma, medulloblastoma and other human tumours. There is mounting clinical evidence that the mutational status of cancer driver genes such as KRAS or IDH1 may influence the risk of venous thromboembolism in patients with colorectal, lung or brain cancers. Notably, single cell sequencing in glioblastoma revealed a remarkable intra-tumoural heterogeneity of cancer cell populations with regard to their individual coagulomes, suggesting a combinatorial and dynamic nature of the global pro-thrombotic phenotype. We suggest that the cellular complexity of specific cancers may define their mechanisms of interactions with the coagulation system, and the risks of thrombosis. Thus, more biologically- based, disease-specific and personalized approaches may be needed to diagnose and manage cancer-related thrombosis.
Collapse
Affiliation(s)
- Nadim Tawil
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | | | - Rayhaan Bassawon
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Radia Johnson
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Esterina D'Asti
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Janusz Rak
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada.
| |
Collapse
|
38
|
Ünlü B, Versteeg HH. Cancer-associated thrombosis: The search for the holy grail continues. Res Pract Thromb Haemost 2018; 2:622-629. [PMID: 30349879 PMCID: PMC6178660 DOI: 10.1002/rth2.12143] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/24/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer patients have an increased risk of developing venous thromboembolism (VTE), a condition that is associated with increased morbidity and mortality. Although risk assessment tools have been developed, it is still very challenging to predict which cancer patients will suffer from VTE. The scope of this review is to summarize and discuss studies focusing on the link between genetic alterations and risk of cancer-associated thrombosis (CAT). Thus far, classical risk factors that contribute to VTE have been tried as risk factors of CAT, with low success. In support, hypercoagulant plasma profiles in patients with CAT differ from those with only VTE, indicating other risk factors that contribute to VTE in cancer. As germline mutations do not significantly contribute to elevated risk of VTE, somatic mutations in tumors may significantly associate with and contribute to CAT. As it is very time-consuming to investigate each and every mutation, an unbiased approach is warranted. In this light we discuss our own recent unbiased proof-of-principle study using RNA sequencing in isolated colorectal cancer cells. Our work has uncovered candidate genes that associate with VTE in colorectal cancer, and these gene profiles associated with VTE more significantly than classical parameters such as platelet counts, D-dimer, and P-selectin levels. Genes associated with VTE could be linked to pathways being involved in coagulation, inflammation and methionine degradation. We conclude that tumor cell-specific gene expression profiles and/or mutational status has superior potential as predictors of VTE in cancer patients.
Collapse
Affiliation(s)
- Betül Ünlü
- Department of Internal MedicineEinthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Henri H. Versteeg
- Department of Internal MedicineEinthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
39
|
Dou F, Li H, Zhu M, Liang L, Zhang Y, Yi J, Zhang Y. Association between oncogenic status and risk of venous thromboembolism in patients with non-small cell lung cancer. Respir Res 2018; 19:88. [PMID: 29743116 PMCID: PMC5944093 DOI: 10.1186/s12931-018-0791-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/26/2018] [Indexed: 01/06/2023] Open
Abstract
Background Preclinical data suggest that oncogene (EGFR and KRAS) events regulate tumor procoagulant activity. However, few studies have prospectively investigated the clinical relevance between the presence of EGFR or KRAS mutations and occurrence of venous thromboembolism(VTE) in patients with non-small cell lung cancer (NSCLC). Methods A total of 605 Chinese patients with newly diagnosed NSCLC were included and were followed for a maximum period of 4.5 years. EGFR and KRAS mutations were determined by amplification refractory mutation system polymerase chain reaction method at inclusion. The main outcome was objectively confirmed VTE. Results Of the 605 patients, 40.3% (244) had EGFR mutations and 10.2% (62) of patients had KRAS mutations. In multivariable analysis including age, sex, tumor histology, tumor stage, performance status, EGFR and KRAS status, EGFR wild-type (sub-distribution hazard ratio 1.81, 95% confidence interval 1.07–3.07) were associated with the increased risk of VTE. In competing risk analysis, the probability of developing VTE was 8.3% in those with and 13.2% in those without EGFR mutations after 1 year; after 2 years, the corresponding risks were 9.7 and 15.5% (Gray test P = 0.047). Conclusions EGFR mutations have a negative association with the risk of VTE in Chinese patients with NSCLC.
Collapse
Affiliation(s)
- Feifei Dou
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China
| | - Huiqiao Li
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China
| | - Lirong Liang
- Department of Clinical Epidemiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yuan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China
| | - Jiawen Yi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, 100020, China.
| |
Collapse
|
40
|
Shen Q, Dong XQ, Zhou JY. Long-term Survival Associated with Crizotinib in a Lung Cancer Patient with a Pulmonary Artery Embolism. Chin Med J (Engl) 2018; 131:111-112. [PMID: 29271393 PMCID: PMC5754949 DOI: 10.4103/0366-6999.221265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Qian Shen
- Department of Respiratory Diseases, The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiao-Qi Dong
- Department of Respiratory Diseases, The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Jian-Ying Zhou
- Department of Respiratory Diseases, The First Affiliated Hospital of College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
41
|
Shen Q, Dong X, Tang X, Zhou J. Risk factors and prognosis value of venous thromboembolism in patients with advanced non-small cell lung cancer: a case-control study. J Thorac Dis 2017; 9:5068-5074. [PMID: 29312712 DOI: 10.21037/jtd.2017.11.116] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Venous thromboembolism (VTE) is the well-known complication of cancer, especially among lung cancer patients. The objective of this study is to identify the incidence, risk factors and prognosis of VTE in the patients with non-small cell lung cancer (NSCLC) in an advanced or metastatic stage (stage III or IV). We hypothesized that NSCLC patients associated with VTE may lead to poor prognosis. Methods From 2012 to 2015, we retrospectively investigated 1,560 patients diagnosed with lung cancer in stage III or IV. Each VTE patient was matched with three control patients according to gender, age, pathology, clinical stage and the diagnosed time. Univariate and multivariate analyses were used in the study. Results Among the 1,560 patients in our study, 32 (2.0%) developed VTE in locally advanced or metastatic stage lung cancer. Weight loss, serous effusion, the absence of the EGFR mutation, poor performance status (PS), hypoalbuminemia, hyponatremia, long prothrombin time (PT), and elevated levels of C-reaction-protein (CRP) and D-dimer were found to be associated with an increased risk of VTE by univariate analyses. The multivariate analyses revealed that weight loss, poor PS, increased CRP and long PT were significantly related to VTE. Overall survival (OS) from diagnosis was significantly shorter in patients with VTE (medians 14.2 vs. 24.4 months; P<0.001). Conclusions The incidence of VTE was high among advanced NSCLC patients, particularly for those with weight loss, poor PS, increased CRP and long PT. Moreover, the patients with VTE complications may have a poorer prognosis.
Collapse
Affiliation(s)
- Qian Shen
- Department of Respiratory Disease, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaoqi Dong
- Department of Respiratory Disease, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xiaoping Tang
- Department of Respiratory Disease, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianying Zhou
- Department of Respiratory Disease, First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
42
|
Unruh D, Schwarze SR, Khoury L, Thomas C, Wu M, Chen L, Chen R, Liu Y, Schwartz MA, Amidei C, Kumthekar P, Benjamin CG, Song K, Dawson C, Rispoli JM, Fatterpekar G, Golfinos JG, Kondziolka D, Karajannis M, Pacione D, Zagzag D, McIntyre T, Snuderl M, Horbinski C. Mutant IDH1 and thrombosis in gliomas. Acta Neuropathol 2016; 132:917-930. [PMID: 27664011 PMCID: PMC5640980 DOI: 10.1007/s00401-016-1620-7] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 10/21/2022]
Abstract
Mutant isocitrate dehydrogenase 1 (IDH1) is common in gliomas, and produces D-2-hydroxyglutarate (D-2-HG). The full effects of IDH1 mutations on glioma biology and tumor microenvironment are unknown. We analyzed a discovery cohort of 169 World Health Organization (WHO) grade II-IV gliomas, followed by a validation cohort of 148 cases, for IDH1 mutations, intratumoral microthrombi, and venous thromboemboli (VTE). 430 gliomas from The Cancer Genome Atlas were analyzed for mRNAs associated with coagulation, and 95 gliomas in a tissue microarray were assessed for tissue factor (TF) protein. In vitro and in vivo assays evaluated platelet aggregation and clotting time in the presence of mutant IDH1 or D-2-HG. VTE occurred in 26-30 % of patients with wild-type IDH1 gliomas, but not in patients with mutant IDH1 gliomas (0 %). IDH1 mutation status was the most powerful predictive marker for VTE, independent of variables such as GBM diagnosis and prolonged hospital stay. Microthrombi were far less common within mutant IDH1 gliomas regardless of WHO grade (85-90 % in wild-type versus 2-6 % in mutant), and were an independent predictor of IDH1 wild-type status. Among all 35 coagulation-associated genes, F3 mRNA, encoding TF, showed the strongest inverse relationship with IDH1 mutations. Mutant IDH1 gliomas had F3 gene promoter hypermethylation, with lower TF protein expression. D-2-HG rapidly inhibited platelet aggregation and blood clotting via a novel calcium-dependent, methylation-independent mechanism. Mutant IDH1 glioma engraftment in mice significantly prolonged bleeding time. Our data suggest that mutant IDH1 has potent antithrombotic activity within gliomas and throughout the peripheral circulation. These findings have implications for the pathologic evaluation of gliomas, the effect of altered isocitrate metabolism on tumor microenvironment, and risk assessment of glioma patients for VTE.
Collapse
Affiliation(s)
- Dusten Unruh
- Department of Neurosurgery, Northwestern University, Tarry 2-705, 300 East Superior Street, Chicago, IL, 60611, USA
| | | | - Laith Khoury
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
| | - Cheddhi Thomas
- Department of Pathology, New York University, New York, NY, USA
| | - Meijing Wu
- Department of Neurosurgery, Northwestern University, Tarry 2-705, 300 East Superior Street, Chicago, IL, 60611, USA
| | - Li Chen
- Department of Biostatistics, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Rui Chen
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Yinxing Liu
- Department of Pathology, University of Kentucky, Lexington, KY, USA
| | | | - Christina Amidei
- Department of Neurosurgery, Northwestern University, Tarry 2-705, 300 East Superior Street, Chicago, IL, 60611, USA
| | - Priya Kumthekar
- Department of Neurology, Northwestern University, Chicago, IL, USA
| | | | | | | | | | | | - John G Golfinos
- Department of Neurosurgery, New York University, New York, NY, USA
| | | | | | - Donato Pacione
- Department of Neurosurgery, New York University, New York, NY, USA
| | - David Zagzag
- Department of Pathology, New York University, New York, NY, USA
- Department of Neurosurgery, New York University, New York, NY, USA
| | - Thomas McIntyre
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Matija Snuderl
- Department of Pathology, New York University, New York, NY, USA
| | - Craig Horbinski
- Department of Neurosurgery, Northwestern University, Tarry 2-705, 300 East Superior Street, Chicago, IL, 60611, USA.
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
43
|
Alexander M, Burbury K. A systematic review of biomarkers for the prediction of thromboembolism in lung cancer - Results, practical issues and proposed strategies for future risk prediction models. Thromb Res 2016; 148:63-69. [PMID: 27815968 DOI: 10.1016/j.thromres.2016.10.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/13/2016] [Accepted: 10/24/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION This review aimed to identify candidate biomarkers for the prediction of thromboembolism (TE) in lung cancer. MATERIALS AND METHODS Systematic review of publications indexed in PubMed or EMBASE databases in the past 5years (01/05/2011-01/05/2016) which evaluated baseline and/or longitudinal biomarker measurements as a predictor of subsequent TE (venous and arterial) in lung cancer patients. RESULTS Of 1105 studies identified, 18 fulfilled predefined inclusion criteria: 6 prospective and 12 retrospective. The 18 studies included 11,262 patients and 36 unique biomarkers. The combined TE rate was 7% (741/10,854), increasing to 11% (294/2612) within prospective studies. All biomarker measurements were baseline only, with no longitudinal assessment reported. The most frequently investigated biomarkers were tumour-related driver mutations, D-dimer, haemoglobin, white cell, and platelet count; as well as biomarker combinations previously used in risk prediction models, such as Khorana risk score. Biomarker thresholds rather than continuous variable analyses were generally applied, however thresholds were not consistent across studies. D-dimer and epidermal growth factor receptor mutation were the strongest and most reproducible predictors of TE. CONCLUSION An important limitation is the lack of prospective data across specific subpopulations of cancer, with correlative, and preferably longitudinal, biomarker assessments. This would provide insight into the pathophysiology, allow patient profiling, and the development of personalised decision-making tools that can be used real-time and throughout the course of the patients' journey, for targeted, risk-adaptive preventative strategies.
Collapse
Affiliation(s)
- Marliese Alexander
- Department of Epidemiology and Preventative Medicine Monash University, Commercial Road Melbourne Australia; Departments of Pharmacy and Haematology Peter MacCallum Cancer Centre, Grattan Street Melbourne Australia.
| | - Kate Burbury
- Department of Haematology Peter MacCallum Cancer Centre, Grattan Street Melbourne Australia
| |
Collapse
|