Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
2.
Clin Appl Thromb Hemost ; 28: 10760296221097969, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35733370

RESUMO

Thrombotic and hemorrhagic complications are related to a significant rate of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs), they are therefore called "thrombohemorrhagic" syndromes. Several clinical factors, such as age and presence of cardiovascular comorbidities are responsible for thrombotic complications. High blood counts, platelet alterations, presence of JAK2 mutation and possibly of other CHIP mutations such as TET2, DNMT3A, and ASXL1, procoagulant microparticles, NETs formation, endothelial activation and neo-angiogenesis are some of the parameters accounting for hypercoagulability in patients with myeloproliferative neoplasms. Bleeding complications emerge as a result of platelet exhaustion. They can be also linked to a functional deficiency of von Willebrand factor, when platelet counts rise above 1000G/L. The mainstay of management consists on preventing hemostatic complications, by antiplatelet and/or anticoagulant treatment and myelosuppressive agents in high-risk patients.Circumstances related to a high thrombohemorrhagic risk, such as pregnancy and the perioperative period, prompt for specific management with regards to anticoagulation and myelosuppression treatment type. In order to apply a patient-specific treatment strategy, there is a need for a risk score assessment tool encompassing clinical parameters and hemostasis biomarkers.


Assuntos
Transtornos Mieloproliferativos , Neoplasias , Trombose , Plaquetas , Feminino , Hemorragia/terapia , Humanos , Transtornos Mieloproliferativos/complicações , Transtornos Mieloproliferativos/genética , Neoplasias/complicações , Gravidez , Trombose/complicações
3.
TH Open ; 6(2): e89-e95, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35498378

RESUMO

Background Cancer-associated thrombosis (CAT) is the second cause of mortality after cancer itself. CAT is underestimated as a health challenge among oncologists, whereas the levels of awareness among patients and the public have not been systematically assessed and followed in the European Union countries. Aim The Prospective Risk Assessment and Management of Patient with CAT (ROADMAP-CAT) Awareness study is an investigator-initiated, descriptive and nonexperimental study with a cross-sectional design and it explores CAT risk awareness among cancer patients and the general public in Greece to provide an impetus for health policy interventions and a benchmark against which impact of any future interventions may be assessed. Methods A total of 1,003 participants aged above 18 years were contacted by phone after random selection from the national telephone catalogue. Participation was voluntary and completely anonymous, and a structured questionnaire was used to elicit responses. Data were analyzed using IBM SPSS version 25. Results Among respondents, almost one-third (32.3%) reported CAT awareness, while only one in five (21.7%) were aware of the signs and symptoms of venous thromboembolism (VTE). Among patients with a personal history of cancer or of VTE, 47 and 58%, respectively, were aware of CAT risk. Of those aware of the association, 35.2% identified their treating physician as the main source of information. The level of awareness did not significantly differ by responders' demographics. Conclusion The ROADMAP-CAT Awareness study revealed very low levels of awareness on CAT and VTE risk both among the general public and cancer patients in Greece. Awareness of the signs and symptoms of VTE was also particularly low. Treating physicians are not actively engaging in educating their patients about CAT. Public awareness of the increased risk of VTE among cancer patients is critical to prevent and diagnose the disease early. It is imperative that a structured campaign supports medical professionals to take the time to increase awareness and educate their patients on this matter if to improve morbidity and mortality of cancer patients.

4.
Thromb Haemost ; 122(10): 1662-1672, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35483884

RESUMO

Hepatectomy-induced coagulation disturbances have been well studied over the past decade. Cumulative evidence supports the superiority of global coagulation analysis compared with conventional coagulation tests (i.e., prothrombin time or activated partial thromboplastin time) for clinical decision making. Cancer, however, represents an acquired prothrombotic state and liver resection for cancer deserves a more thorough investigation. This prospective observational study was conducted to assess the perioperative coagulation status of patients undergoing major hepatectomies for primary or metastatic hepatic malignancy. Patients were followed up to the 10th post-operative day by serial measurements of conventional coagulation tests, plasma levels of coagulation factors, and thrombin generation assay parameters. An abnormal coagulation profile was detected at presentation and included elevated FVIII levels, decreased levels of antithrombin, and lag time prolongation in thrombin generation. Serial hematological data demonstrated increased Von Willebrand factor, FVIII, D-dimer, fibrinogen and decreased levels of natural anticoagulant proteins in the early post-operative period predisposing to a hyper-coagulable state. The ratio of the anticoagulant protein C to the procoagulant FVIII was low at baseline and further declined post-operatively, indicating a prothrombotic state. Though no bleeding complications were reported, one patient experienced pulmonary embolism while under thromboprophylaxis. Overall, patients with hepatic carcinoma presenting for elective major hepatectomy may have baseline malignancy-associated coagulation disturbances, aggravating the hyper-coagulable state documented in the early post-operative period.


Assuntos
Neoplasias , Tromboembolia Venosa , Anticoagulantes , Antitrombinas , Fatores de Coagulação Sanguínea/metabolismo , Testes de Coagulação Sanguínea , Fibrinogênio/metabolismo , Hepatectomia/efeitos adversos , Humanos , Fígado/metabolismo , Proteína C , Trombina/metabolismo , Fator de von Willebrand/metabolismo
5.
Clin Appl Thromb Hemost ; 28: 1076029620939181, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35187959

RESUMO

Venous thromboembolism (VTE) is a multifactorial disease that can possibly affect any part of venous circulation. The risk of VTE increases by about 2 fold in pregnant women and VTE is one of the major causes of maternal morbidity and mortality. For decades superficial vein thrombosis (SVT) has been considered as benign, self-limiting condition, primarily local event consequently being out of scope of well conducted epidemiological and clinical studies. Recently, the approach on SVT has significantly changed considering that prevalence of lower limb SVT is twice higher than both deep vein thrombosis (DVT) and pulmonary embolism (PE). The clinical severity of SVT largely depends on the localization of thrombosis, when it concerns the major superficial vein vessels of the lower limb and particularly the great saphenous vein. If untreated or inadequately treated, SVT can potentially cause DVT or PE. The purpose of this review is to discuss the complex interconnection between SVT and risk factors in pregnancy and to provide evidence-based considerations, suggestions, and recommendations for the diagnosis and treatment of this precarious and delicate clinical entity.


Assuntos
Anticoagulantes/uso terapêutico , Trombose Venosa/tratamento farmacológico , Trombose Venosa/prevenção & controle , Anticoagulantes/administração & dosagem , Anticoagulantes/efeitos adversos , Península Balcânica , Feminino , Humanos , Gravidez , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Risco
6.
Clin Appl Thromb Hemost ; 28: 10760296211056648, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35167393

RESUMO

The progress in the development of various vaccine platforms against SARS-CoV-2 have been rather remarkable owing to advancement in molecular and biologic sciences. Most of the current vaccines and those in development focus on targeting the viral spike proteins by generating antibodies of varying spectrum. These vaccines represent a variety of platforms including whole virus vaccines, viral vector vaccines, nucleic acid vaccines representing RNA, DNA, and their hybrid forms.The therapeutic efficacy of these vaccines varies owing to their pharmacodynamic individualities. COVID-19 variants are capable of inducing different pathologic responses and some of which may be resistant to antibodies generated by current vaccines. The current clinical use of these vaccines has been through emergency use authorization until recently. Moreover, the efficacy and safety of these vaccines have been tested in substantial numbers of individuals but studies in special populations that better reflect the global population are pending results. These specialized populations include young children, immunocompromised patients, pregnant individuals, and other specialized groups. Combination approaches, molecularly modified vaccination approaches, and vaccines conferring longer periods of immunity are being currently being investigated, as well as pharmacovigilance studies.The continual transformation of SARS-CoV-2 and its variants are of concern along with the breakthrough infections. These considerations pose new challenges for the development of vaccination platforms. For this purpose, booster doses, combination vaccine approaches, and other modalities are being discussed. This review provides an updated account of currently available vaccines and those in advanced development with reference to their composition and mechanisms of action.A discussion on the use of vaccines in special populations including immunocompromised patients, pregnant women and other specialized populations are also included.


Assuntos
Vacinas contra COVID-19/farmacologia , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Desenvolvimento de Vacinas/métodos , Adolescente , COVID-19/imunologia , COVID-19/virologia , Vacinas contra COVID-19/efeitos adversos , Vacinas contra COVID-19/uso terapêutico , Criança , Feminino , Humanos , Hospedeiro Imunocomprometido , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/prevenção & controle , Complicações Infecciosas na Gravidez/virologia
8.
Thromb Haemost ; 121(8): 992-1007, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34169495

RESUMO

BACKGROUND: One year after the declaration of the coronavirus disease 2019 (COVID-19) pandemic by the World Health Organization (WHO) and despite the implementation of mandatory physical barriers and social distancing, humanity remains challenged by a long-lasting and devastating public health crisis. MANAGEMENT: Non-pharmacological interventions (NPIs) are efficient mitigation strategies. The success of these NPIs is dependent on the approval and commitment of the population. The launch of a mass vaccination program in many countries in late December 2020 with mRNA vaccines, adenovirus-based vaccines, and inactivated virus vaccines has generated hope for the end of the pandemic. CURRENT ISSUES: The continuous appearance of new pathogenic viral strains and the ability of vaccines to prevent infection and transmission raise important concerns as we try to achieve community immunity against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and its variants. The need of a second and even third generation of vaccines has already been acknowledged by the WHO and governments. PERSPECTIVES: There is a critical and urgent need for a balanced and integrated strategy for the management of the COVID-19 outbreaks organized on three axes: (1) Prevention of the SARS-CoV-2 infection, (2) Detection and early diagnosis of patients at risk of disease worsening, and (3) Anticipation of medical care (PDA). CONCLUSION: The "PDA strategy" integrated into state policy for the support and expansion of health systems and introduction of digital organizations (i.e., telemedicine, e-Health, artificial intelligence, and machine-learning technology) is of major importance for the preservation of citizens' health and life world-wide.


Assuntos
COVID-19/epidemiologia , COVID-19/prevenção & controle , Saúde Pública , COVID-19/diagnóstico , Teste para COVID-19/métodos , Vacinas contra COVID-19/uso terapêutico , Gerenciamento Clínico , Humanos , Programas de Imunização/métodos , Pandemias/prevenção & controle , Saúde Pública/métodos , Medição de Risco , SARS-CoV-2/isolamento & purificação
9.
Clin Appl Thromb Hemost ; 27: 10760296211021498, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34060379

RESUMO

Today the coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has become a global health problem. After more than a year with the pandemic, although our knowledge has progressed on COVID-19, there are still many unknowns in virological, pathophysiological and immunological aspects. It is obvious that the most efficient solution to end this pandemic are safe and efficient vaccines. This manuscript summarizes the pathophysiological and thrombotic features of COVID-19 and the safety and efficacy of currently approved COVID-19 vaccines with an aim to clarify the recent concerns of thromboembolic events after COVID-19 vaccination. The influx of newer information is rapid, requiring periodic updates and objective assessment of the data on the pathogenesis of COVID-19 variants and the safety and efficacy of currently available vaccines.


Assuntos
Vacinas contra COVID-19/efeitos adversos , COVID-19/prevenção & controle , SARS-CoV-2 , Trombose/etiologia , Ad26COVS1 , Autoanticorpos/biossíntese , Vacina BNT162 , COVID-19/epidemiologia , COVID-19/fisiopatologia , Vacinas contra COVID-19/genética , Vacinas contra COVID-19/imunologia , ChAdOx1 nCoV-19 , Ensaios Clínicos como Assunto , Coagulação Intravascular Disseminada/epidemiologia , Coagulação Intravascular Disseminada/etiologia , Aprovação de Drogas , Feminino , Vetores Genéticos , Glicosaminoglicanos/imunologia , Humanos , Masculino , Modelos Cardiovasculares , Pandemias/prevenção & controle , Fator Plaquetário 4/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Segurança , Trombose dos Seios Intracranianos/epidemiologia , Trombose dos Seios Intracranianos/etiologia , Trombose/epidemiologia , Trombose/fisiopatologia , Vacinas de Produtos Inativados/efeitos adversos , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas de mRNA
10.
Thromb Haemost ; 121(8): 982-991, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33946120

RESUMO

Historically, the vaccination strategies developed in the second half of the 20th century have facilitated the eradication of infectious diseases. From the onset of COVID-19 pandemic to the end of April 2021, more than 150 million cases and 3 million deaths were documented worldwide with disruption of the economic and social activity, and with devastating material, physical, and psychological consequences. Reports of unusual and severe thrombotic events, including cerebral and splanchnic venous thrombosis and other autoimmune adverse reactions, such as immune thrombocytopenia or thrombotic microangiopathies in connection with some of the SARS-CoV-2 vaccines, have caused a great deal of concern within the population and the medical community. This report is intended to provide practical answers following an overview of our knowledge on these thrombotic events that are extremely rare but have serious consequences. Vaccine hesitancy threatens to reverse the progress made in controlling vaccine-preventable diseases. These adverse events must be put into perspective with an objective analysis of the facts and the issues of the vaccination strategy during this SARS-CoV-2 pandemic. Health care professionals remain the most pertinent advisors and influencers regarding vaccination decisions; they have to be supported to provide reliable and credible information on vaccines. We need to inform, reassure, and support our patients when the prescription is made. Facing these challenges and observations, a panel of experts express their insights and propose a tracking algorithm for vaccinated patients based on a 10-point guideline for decision-making on what to do and not to do.


Assuntos
Vacinas contra COVID-19/efeitos adversos , COVID-19/prevenção & controle , Púrpura Trombocitopênica Idiopática/etiologia , Trombose/etiologia , Vacinas contra COVID-19/uso terapêutico , Tomada de Decisão Clínica , Hemorragia/etiologia , Humanos , Fatores de Risco , Trombocitopenia/etiologia , Vacinação/efeitos adversos , Vacinação/métodos
11.
Infection ; 49(3): 559-562, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33393065

RESUMO

Severe 2019 novel coronavirus infectious disease (COVID-19) with pneumonia is associated with high rates of admission to the intensive care unit (ICU). Bacterial coinfection has been reported to be rare. We aimed at describing the rate of bacterial coinfection in critically ill adult patients with severe COVID-19 pneumonia. All the patients with laboratory-confirmed severe COVID-19 pneumonia admitted to the ICU of Tenon University-teaching hospital, from February 22 to May 7th, 2020 were included. Respiratory tract specimens were obtained within the first 48 h of ICU admission. During the study period, 101 patients were referred to the ICU for COVID-19 with severe pneumonia. Most patients (n = 83; 82.2%) were intubated and mechanically ventilated on ICU admission. Overall, 20 (19.8%) respiratory tract specimens obtained within the first 48 h. Staphylococcus aureus was the main pathogen identified, accounting for almost half of the early-onset bacterial etiologies. We found a high prevalence of early-onset bacterial coinfection during severe COVID-19 pneumonia, with a high proportion of S. aureus. Our data support the current WHO guidelines for the management of severe COVID-19 patients, in whom antibiotic therapy directed to respiratory pathogens is recommended.


Assuntos
Infecções Bacterianas/epidemiologia , COVID-19/epidemiologia , Coinfecção/epidemiologia , Idoso , Antibacterianos/uso terapêutico , Bactérias/classificação , Bactérias/isolamento & purificação , Infecções Bacterianas/diagnóstico , Infecções Bacterianas/tratamento farmacológico , COVID-19/diagnóstico , Coinfecção/diagnóstico , Coinfecção/tratamento farmacológico , Estado Terminal , Feminino , Hospitais Universitários , Humanos , Unidades de Terapia Intensiva , Masculino , Pessoa de Meia-Idade , Prevalência , SARS-CoV-2/isolamento & purificação , Staphylococcus aureus/isolamento & purificação , Tratamento Farmacológico da COVID-19
12.
Clin Appl Thromb Hemost ; 26: 1076029620983466, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33372544

RESUMO

The prevalence of thrombosis in lymphoma patients is reportedly high and ranges from 3-10%. Vascular malfunction and inflammatory processes further contribute to the thrombotic activation process in these patients. Andexanet alfa (AA) is an antidote for factor Xa inhibitors and its usage has been reported with thrombotic complications. This study was designed to compare the effect of AA on the thrombin generation (TG) potential. Blood samples from 78 patients with confirmed diagnosis of non-Hodgkin lymphoma (NHL), Hodgkin lymphoma (HL) and Chronic lymphocytic leukemia (CLL) were collected from the University of Belgrade Clinic, Serbia. Normal human plasma (NHP) was used for referencing purposes. Individual samples were supplemented with AA at 100 ug/ml. TG studies were carried out using a commercially available fluorogenic substrate method. TG parameters such as peak thrombin (PT), lag time (LT) and area under the curve (AUC) were compiled. Cumulatively, lymphoma patients showed an increase in LT compared to NHP which decreases with AA. The PT and AUC levels were decreased compared to NHP and increases with AA. Upon sub-grouping of lymphoma patients, PT levels for all sub-groups were increased with AA. The AUC values increased for HL and NHL and decreased for CLL with AA. Variations in lag time were noted in all 3 sub-groups. Lymphoma represents a heterogenous group of patients where both the hypercoagulable state and inflammatory responses simultaneously occur. Increased thrombin generation in post AA supplemented samples suggest that the use of this agent may potentially be associated with thrombotic complications.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Fator Xa/farmacologia , Doença de Hodgkin/sangue , Leucemia Linfocítica Crônica de Células B/sangue , Linfoma não Hodgkin/sangue , Proteínas Recombinantes/farmacologia , Testes de Coagulação Sanguínea , Fator Xa/efeitos adversos , Feminino , Doença de Hodgkin/complicações , Humanos , Leucemia Linfocítica Crônica de Células B/complicações , Linfoma não Hodgkin/complicações , Masculino , Proteínas Recombinantes/efeitos adversos , Trombina/análise , Trombose/sangue , Trombose/induzido quimicamente , Trombose/etiologia
13.
Clin Appl Thromb Hemost ; 26: 1076029620964590, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33284037

RESUMO

In patients with stable coronary artery disease (CAD) blood hypercoagulability figures among factors leading to thrombosis. Tissue factor (TF) exposure at ruptured plaque initiates blood coagulation and hypercoagulability is responsible for thrombus formation. Early identification of patients eligible for angiography is a challenging issue for effective prevention of ACS. This pilot study aimed to identify biomarkers of hypercoagulability that can be prospectively used in risk assessment tools for the evaluation of CAD severity. Biomarkers of hypercoagulability could be a used for the evaluation of CAD severity. Platelet-poor plasma from 66 patients who were referred to coronary angiography was assessed for thrombin generation, phospholipid-dependent clotting time (Procoag-PPL ® ) and D-Dimers, and evaluated against atherosclerotic burden. Patients with CAD, as compared to controls, showed attenuated thrombin generation lag time: 4.7 (3.8-5.4) min versus 2.5 (2.1-2.9) min; p < 0.0001, shorter Procoag-PPL® clotting time 55.0(32-66) s versus 62.8 (42-85) s; p = 0.001), and higher D-Dimer levels 0.509 (0.27-2.58) µg/ml versus 0.309 (0.23-0.39) µg/ml; p = 0.038. Multivariate logistic regression model showed excellent discriminatory value in predicting CAD severity. The ROADMAP-CAD study showed that the Procoag-PPL® clotting time and thrombin Peak are informative for the the burden of the coronary atherosclerotic disease. The clinical relevance of this observation in the development of a new clinic-biological risk assessment model for early diagnosis of severe CAD has to be examined in a prospective study.


Assuntos
Doença da Artéria Coronariana/sangue , Trombofilia/sangue , Idoso , Biomarcadores/sangue , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Doença da Artéria Coronariana/diagnóstico , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Estudos Prospectivos , Medição de Risco , Trombofilia/diagnóstico
14.
Thromb Haemost ; 120(12): 1680-1690, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32961572

RESUMO

The prospective observational cohort study COMPASS-COVID-19 aimed to develop a risk assessment model for early identification of hospitalized COVID-19 patients at risk for worsening disease. Patients with confirmed COVID-19 (n = 430) hospitalized between March 18 and April 21, 2020 were divided in derivation (n = 310) and validation (n = 120) cohorts. Two groups became evident: (1) good prognosis group (G-group) with patients hospitalized at the conventional COVID-19 ward and (2) Worsening disease group (W-group) with patients admitted to the intensive care unit (ICU) from the emergency departments. The study end point was disease worsening (acute respiratory failure, shock, myocardial dysfunction, bacterial or viral coinfections, and acute kidney injury) requiring ICU admission. All patients were routinely evaluated for full blood count, prothrombin time, fibrinogen, D-dimers, antithrombin (AT), and protein C activity. Data from the first hospitalization day at the conventional ward or the ICU were analyzed. Cardiovascular risk factors and comorbidities were routinely registered. Obesity, hypertension, diabetes and male gender, increased fibrinogen and D-dimers, thrombocytopenia, AT deficiency, lymphopenia, and an International Society on Thrombosis and Haemostasis (ISTH) score for compensated disseminated intravascular coagulation score (cDIC-ISTH) ≥5 were significant risk factors for worsening disease. The COMPASS-COVID-19 score was derived from multivariate analyses and includes obesity, gender, hemoglobin, lymphocyte, and the cDIC-ISTH score (including platelet count, prothrombin time, D-dimers, AT, and protein C levels). The score has a very good discriminating capacity to stratify patients at high and low risk for worsening disease, with an area under the receiver operating characteristic curve value of 0.77, a sensitivity of 81%, and a specificity of 60%. Application of the COMPASS-COVID-19 score at the validation cohort showed 96% sensitivity. The COMPASS-COVID-19 score is an accurate clinical decision-making tool for an easy identification of COVID-19 patients being at high risk for disease worsening.


Assuntos
COVID-19/epidemiologia , SARS-CoV-2/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Progressão da Doença , Feminino , França/epidemiologia , Grécia/epidemiologia , Hospitalização/estatística & dados numéricos , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Prospectivos , Medição de Risco , Adulto Jovem
15.
Thromb Haemost ; 120(12): 1597-1628, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32920811

RESUMO

COVID-19 is also manifested with hypercoagulability, pulmonary intravascular coagulation, microangiopathy, and venous thromboembolism (VTE) or arterial thrombosis. Predisposing risk factors to severe COVID-19 are male sex, underlying cardiovascular disease, or cardiovascular risk factors including noncontrolled diabetes mellitus or arterial hypertension, obesity, and advanced age. The VAS-European Independent Foundation in Angiology/Vascular Medicine draws attention to patients with vascular disease (VD) and presents an integral strategy for the management of patients with VD or cardiovascular risk factors (VD-CVR) and COVID-19. VAS recommends (1) a COVID-19-oriented primary health care network for patients with VD-CVR for identification of patients with VD-CVR in the community and patients' education for disease symptoms, use of eHealth technology, adherence to the antithrombotic and vascular regulating treatments, and (2) close medical follow-up for efficacious control of VD progression and prompt application of physical and social distancing measures in case of new epidemic waves. For patients with VD-CVR who receive home treatment for COVID-19, VAS recommends assessment for (1) disease worsening risk and prioritized hospitalization of those at high risk and (2) VTE risk assessment and thromboprophylaxis with rivaroxaban, betrixaban, or low-molecular-weight heparin (LMWH) for those at high risk. For hospitalized patients with VD-CVR and COVID-19, VAS recommends (1) routine thromboprophylaxis with weight-adjusted intermediate doses of LMWH (unless contraindication); (2) LMWH as the drug of choice over unfractionated heparin or direct oral anticoagulants for the treatment of VTE or hypercoagulability; (3) careful evaluation of the risk for disease worsening and prompt application of targeted antiviral or convalescence treatments; (4) monitoring of D-dimer for optimization of the antithrombotic treatment; and (5) evaluation of the risk of VTE before hospital discharge using the IMPROVE-D-dimer score and prolonged post-discharge thromboprophylaxis with rivaroxaban, betrixaban, or LMWH.


Assuntos
COVID-19/diagnóstico , Cardiologia , Doenças Cardiovasculares/diagnóstico , SARS-CoV-2/fisiologia , Anticoagulantes/uso terapêutico , COVID-19/epidemiologia , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/epidemiologia , Europa (Continente) , Heparina de Baixo Peso Molecular/uso terapêutico , Humanos , Inflamação , Guias de Prática Clínica como Assunto , Fatores de Risco , Rivaroxabana/uso terapêutico , Sociedades Médicas , Trombofilia , Trombose , Tratamento Farmacológico da COVID-19
16.
Thromb Res ; 191 Suppl 1: S50-S57, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32736779

RESUMO

A B S T R A C T Important progress has been made in the development of risk assessment models (RAM) for the identification of outpatients on anticancer treatment at risk of venous thromboembolism (VTE). Since the breakthrough publication of the original Khorana risk score (KRS) more than 10 years ago, a new generation of KRS-based scores have been developed, including the Vienna Cancer and Thrombosis Study, PROTECHT, CONKO, ONCOTEV, TicOnco and the CATS/MICA score. Among these the CATS/MICA score showed that a simplified score composed of only two calibrated predictors, the type of cancer and the D-dimer levels, offers a user-friendly tool for the evaluation of cancer-associated thrombosis (CAT) risk. The COMPASS-CAT score is the first that introduced a more synthetic approach of risk evaluation by combining cancer-related predictors with patient comorbidity in a score which is designed for the types of cancer frequently seen in the community (i.e. breast, lung colon or ovarian cancers) and has been externally validated in independent studies. The Throly score is registered as part of the same group as it has a similar structure to the COMPASS-CAT score and is applicable in patients with lymphoma. The incorporation of specific biomarkers of hypercoagulability to the RAM for CAT offers the possibility to perform a precision medicine approach in the prevention of CAT. The improvement of RAM for CAT with artificial intelligence methodologies and deep learning techniques is the challenge in the near future.


Assuntos
Neoplasias , Tromboembolia Venosa , Inteligência Artificial , Humanos , Neoplasias/complicações , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Tromboembolia Venosa/etiologia
17.
Am J Hematol ; 95(7): 834-847, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32282949

RESUMO

COVID-19 is a systemic infection with a significant impact on the hematopoietic system and hemostasis. Lymphopenia may be considered as a cardinal laboratory finding, with prognostic potential. Neutrophil/lymphocyte ratio and peak platelet/lymphocyte ratio may also have prognostic value in determining severe cases. During the disease course, longitudinal evaluation of lymphocyte count dynamics and inflammatory indices, including LDH, CRP and IL-6 may help to identify cases with dismal prognosis and prompt intervention in order to improve outcomes. Biomarkers, such high serum procalcitonin and ferritin have also emerged as poor prognostic factors. Furthermore, blood hypercoagulability is common among hospitalized COVID-19 patients. Elevated D-Dimer levels are consistently reported, whereas their gradual increase during disease course is particularly associated with disease worsening. Other coagulation abnormalities such as PT and aPTT prolongation, fibrin degradation products increase, with severe thrombocytopenia lead to life-threatening disseminated intravascular coagulation (DIC), which necessitates continuous vigilance and prompt intervention. So, COVID-19 infected patients, whether hospitalized or ambulatory, are at high risk for venous thromboembolism, and an early and prolonged pharmacological thromboprophylaxis with low molecular weight heparin is highly recommended. Last but not least, the need for assuring blood donations during the pandemic is also highlighted.


Assuntos
Betacoronavirus , Infecções por Coronavirus/sangue , Linfopenia/etiologia , Pneumonia Viral/sangue , Trombofilia/etiologia , Anticoagulantes/uso terapêutico , Biomarcadores , Testes de Coagulação Sanguínea , Doadores de Sangue/provisão & distribuição , Proteína C-Reativa/análise , COVID-19 , Teste para COVID-19 , Técnicas de Laboratório Clínico , Infecções por Coronavirus/complicações , Infecções por Coronavirus/diagnóstico , Síndrome da Liberação de Citocina/sangue , Síndrome da Liberação de Citocina/etiologia , Citocinas/sangue , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/etiologia , Diagnóstico Precoce , Ferritinas/sangue , Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Humanos , Metanálise como Assunto , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/diagnóstico , Risco , SARS-CoV-2 , Trombofilia/sangue , Trombofilia/tratamento farmacológico , Tromboembolia Venosa/sangue , Tromboembolia Venosa/etiologia , Tromboembolia Venosa/prevenção & controle
18.
Thromb Res ; 187: 170-179, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32006891

RESUMO

The endothelium could be a potential target of cancer cell derived extracellular vesicles (CaCe-dEV). We investigated in vitro the effect of CaCe-dEV on the hemostatic balance of endothelial cells. Extracellular vesicles released from pancreas adenocarcinoma cells (BXPC3) or human breast cancer cells (MCF7) were isolated by differential centrifugation. Human umbilical vein endothelial cells (HUVEC) were cultured for 72 h in the presence or absence of CaCe-dEV. Subsequently, they were washed and re-cultivated over three cycles to get daughter cell generations (DG) which were not exposed to CaCe-dEV. Thrombin generation of normal platelet poor plasma (PPP) added in wells carrying HUVEC was assessed by the Calibrated Automated Thrombogram®. Tissue factor activity (TFa) and procoagulant phospholipid clotting time were assessed. Some traces of TFa were displayed by non-exposed HUVEC (0.18 ±â€¯0.03 pM) and their EVs (1.2 ±â€¯1.0 pM). Non-exposed HUVEC did not induce any detectable thrombin generation. BXPC3-dEV displayed significantly higher TFa as compared to MCF7-dEV (45 ±â€¯5 pM versus 4.6 ±â€¯2.3pM respectively; p < 0.05). HUVEC exposed to CaCe-dEV enhanced thrombin generation. BXPC3-dEV induced significantly higher thrombin generation as compared to those exposed to MCF7-dEV. The procoagulant properties of HUVEC, acquired upon exposure to CaCe-dEV were transferred to DG. In conclusion, CaCe-dEV lead to a procoagulant shift of endothelial cells which, upon exposure, display TFa and enhance thrombin generation which is transferred to DG of HUVEC. The potency of CaCe-dEV to induce procoagulant shift of HUVEC depends on the histological type of the cancer cells. The procoagulant shift of endothelial cells which is transferable to DG could be an additional mechanism - together with cancer-induced blood hypercoagulability - in the pathogenesis of cancer associated thrombosis.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , Neoplasias Pancreáticas , Feminino , Humanos , Pâncreas , Trombina , Tromboplastina
19.
TH Open ; 3(4): e340-e347, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31693008

RESUMO

Introduction Hypercoagulability is a common blood alteration in newly diagnosed chemotherapy naïve patients with multiple myeloma. The identification of the procoagulant potential of cancer cells, which is principally related to tissue factor (TF) expression, attracts particular interest. The mechanisms by which myeloma plasma cells (MPCs) activate blood coagulation have been poorly investigated. Aim To identify the principal actors related with MPCs that boost thrombin generation (TG). Methods TF and annexin V expression by MPCs and MPC-derived microparticles (MPC-dMPs) was analyzed by flow cytometry. TF activity (TFa) and TF gene expression were also determined. TG in the presence of MPCs or MPC-dMPs was assessed with the calibrated automated thrombogram assay (CAT) in normal human PPP and in plasma depleted of factor VII or XII. TG was also assessed in plasma spiked with MPCs and MPC-dMPs. Results MPC-dMPs expressed approximately twofold higher levels of TF as compared with MPCs. The TFa expressed by MPC-dMPs was significantly higher compared with that expressed by MPCs. MPCs and MPC-dMPs enhanced TG of human plasma. TG was significantly higher with MPC-dMPs compared with MPCs. Conclusion MPCs indirectly induce blood-borne hypercoagulability through the release of MPC-dMPs rich in TF. Since MPCs, expressing low TFa, represent a weak procoagulant stimulus, the hypercoagulability at the microenvironment could be the resultant of MPC-dMPs rich in TF.

20.
TH Open ; 3(4): e348-e349, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31696158

RESUMO

[This corrects the article DOI: 10.1055/s-0039-1696659.].

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...