Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38546698

RESUMEN

Chimeric antigen receptor (CAR-) T cell therapy causes serious side effects including cytokine release syndrome (CRS). CRS-related coagulopathy is associated with hypofibrinogenemia that is thus far considered the result of disseminated intravascular coagulation (DIC) and liver dysfunction. We investigated incidence and risk factors for hypofibrinogenemia in 41 consecutive adult patients with hematologic malignancies (median age 69 years, range 38-83 years) receiving CAR-T cell therapy between 01/2020 and 05/2023 at the University Medical Center Regensburg. CRS occurred in 93% of patients and was accompanied by hypofibrinogenemia already from CRS grade 1. Yet, DIC and liver dysfunction mainly occurred in severe CRS (≥ grade 3). After an initial increase during CRS, fibrinogen levels dropped after administration of tocilizumab in a dose dependent manner (r = -0.44, p = 0.004). In contrast, patients who did not receive tocilizumab had increased fibrinogen levels. Logistic regression analysis identified tocilizumab as an independent risk factor for hypofibrinogenemia (odds ratio = 486, p < 0.001). We thus hypothesize that fibrinogen synthesis in CRS is upregulated in an interleukin-6-dependent acute phase reaction compensating for CRS-induced consumption of coagulation factors. Tocilizumab inhibits fibrinogen upregulation resulting in prolonged hypofibrinogenemia. These observations provide novel insights into the pathophysiology of hypofibrinogenemia following CAR-T cell therapy and emphasize the need for close fibrinogen monitoring after tocilizumab treatment of CRS.

2.
Acta Haematol ; 145(4): 454-457, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35086107

RESUMEN

Acquired von Willebrand Syndrome (AVWS) is a rare coagulation disorder which can be associated with IgM paraproteinaemia. Recently, recombinant von Willebrand factor (rVWF) has become available for the treatment of bleedings in patients with inherited von Willebrand disease, but experience in patients with AVWS is limited. We report on 2 patients with AVWS with underlying IgM paraproteinaemia with distinct underlying pathomechanisms. In 1 patient, the paraprotein built unspecific complexes with von Willebrand factor (VWF). In the other patient, we were able to detect an IgM antibody against VWF. Bleeding in this patient was successfully treated with rVWF. To our knowledge, this is the first report about the successful use of rVWF in a patient with AVWS with the detection of a VWF-specific antibody.


Asunto(s)
Paraproteinemias , Enfermedades de von Willebrand , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Humanos , Inmunoglobulina M/uso terapéutico , Paraproteinemias/diagnóstico , Enfermedades de von Willebrand/diagnóstico , Enfermedades de von Willebrand/tratamiento farmacológico , Factor de von Willebrand/uso terapéutico
3.
Cytokine ; 120: 192-201, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31100684

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease of complex and still poorly understood etiology. Loss of upper and lower motoneurons results in death within few years after diagnosis. Recent studies have proposed neuroprotective and disease-slowing effects of granulocyte-colony stimulating factor (G-CSF) treatment in ALS mouse models as well as humans. In this study, six ALS patients were monitored up to 3.5 years during continuous high-dose G-CSF administration. Repetitive analyses were performed including blood count parameters, CD34+ hematopoietic stem and progenitor cell (HSPC) and colony forming cell (CFC) counts, serum cytokine levels and leukocyte telomere length. We demonstrate that continuous G-CSF therapy was well tolerated and safe resulting in only mild adverse events during the observation period. However, no mobilization of CD34+ HSPC was detected as compared to baseline values. CFC mobilization was equally low and even a decrease of myeloid precursors was observed in some patients. Assessment of telomere length within ALS patients' leukocytes revealed that G-CSF did not significantly shorten telomeres, while those of ALS patients were shorter compared to age-matched healthy controls, irrespective of G-CSF treatment. During G-CSF stimulation, TNF-alpha, CRP, IL-16, sVCAM-1, sICAM-1, Tie-2 and VEGF were significantly increased in serum whereas MCP-1 levels decreased. In conclusion, our data show that continuous G-CSF treatment fails to increase circulating CD34+ HSPC in ALS patients. Cytokine profiles revealed G-CSF-mediated immunomodulatory and proteolytic effects. Interestingly, despite intense G-CSF stimulation, telomere length was not significantly shortened.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movilización de Célula Madre Hematopoyética , Homeostasis del Telómero , Adulto , Anciano , Esclerosis Amiotrófica Lateral/sangre , Antígenos CD34/metabolismo , Recuento de Células Sanguíneas , Ensayo de Unidades Formadoras de Colonias , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto
4.
Br J Haematol ; 179(4): 586-597, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28961309

RESUMEN

We assessed the safety and efficacy of bortezomib, cyclophosphamide and dexamethasone (VCD) induction therapy in previously untreated multiple myeloma patients. A total of 414 patients received three 21-day cycles of VCD prior to autologous stem-cell transplantation (ASCT). Most common grade ≥3 adverse events were leucopenia (31·4%) and thrombocytopenia (6·8%). The overall response rate (ORR) by investigator-based assessment was 85·4%. Most patients (74%) underwent successful central laboratory-based molecular cytogenetic analysis. No clinically relevant differences in ORR post-induction were seen between patients with or without high-risk cytogenetic abnormalities (86·2% vs. 84·3%). Further follow-up data are available for 113 patients receiving ASCT who were included in a prospective consolidation trial (median follow-up, 55·5 months); median progression-free survival (PFS) was 35·3 months and median overall survival (OS) was not reached. In patients with high-risk versus standard-risk cytogenetics, median PFS was 19·9 vs. 43·6 months (P < 0·0001), and median OS was 54·7 months versus not reached (P = 0·0022). VCD is an effective and tolerable induction regimen; results suggest that VCD induces high response rates independently of cytogenetic risk status, but after long-term follow-up, cytogenetic high risk is associated with markedly reduced PFS and OS post-ASCT.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioterapia de Inducción/métodos , Mieloma Múltiple/tratamiento farmacológico , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Bortezomib/administración & dosificación , Quimioterapia de Consolidación , Ciclofosfamida/administración & dosificación , Citogenética , Dexametasona/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Medición de Riesgo/métodos , Trasplante de Células Madre , Análisis de Supervivencia , Trasplante Autólogo , Adulto Joven
5.
Haematologica ; 101(11): 1407-1416, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27662011

RESUMEN

Myelofibrosis is a myeloproliferative neoplasm that results in cytopenia, bone marrow fibrosis and extramedullary hematopoiesis. Allogeneic hematopoietic stem cell transplantation is the only curative treatment but is associated with a risk of delayed engraftment and graft failure. In this study, patients with myelofibrosis (n=31) and acute myeloid leukemia (n=31) were analyzed for time to engraftment, graft failure and engraftment-related factors. Early and late neutrophil engraftment and late thrombocyte engraftment were significantly delayed in patients with myelofibrosis as compared to acute myeloid leukemia, and graft failure only occurred in myelofibrosis (6%). Only spleen size had a significant influence on engraftment efficiency in myelofibrosis patients. To analyze the cause for the engraftment defect, clearance of hematopoietic stem cells from peripheral blood was measured and immunohistological staining of bone marrow sections was performed. Numbers of circulating CD34+ were significantly reduced at early time points in myelofibrosis patients, whereas CD34+CD38- and colony-forming cells showed no significant difference in clearance. Staining of bone marrow sections for homing proteins revealed a loss of VCAM-1 in myelofibrosis with a corresponding significant increase in the level of soluble VCAM-1 within the peripheral blood. In conclusion, our data suggest that reduced engraftment and graft failure in myelofibrosis patients is caused by an early pooling of CD34+ hematopoietic stem cells in the spleen and a bone marrow homing defect caused by the loss of VCAM-1. Improved engraftment in myelofibrosis might be achieved by approaches that reduce spleen size and cleavage of VCAM-1 in these patients prior to hematopoietic stem cell transplantation.


Asunto(s)
Funcionamiento Retardado del Injerto/etiología , Trasplante de Células Madre Hematopoyéticas/métodos , Mielofibrosis Primaria/terapia , Bazo/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Adulto , Anciano , Aloinjertos , Femenino , Rechazo de Injerto/etiología , Humanos , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Mielofibrosis Primaria/complicaciones
7.
Cytokine ; 67(1): 21-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24680478

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive neuronal disease resulting in a loss of the upper and lower motor neurons and subsequent death within three to four years after diagnosis. Mouse models and preliminary human exposure data suggest that the treatment with granulocyte-colony stimulating factor (G-CSF) has neuro-protective effects and may delay ALS progression. As data on long-term administration of G-CSF in patients with normal bone marrow (BM) function are scarce, we initiated a compassionate use program including 6 ALS patients with monthly G-CSF treatment cycles. Here we demonstrate that G-CSF injection was safe and feasible throughout our observation period up to three years. Significant decrease of mobilization efficiency occurred in one patient and a loss of immature erythroid progenitors was observed in all six patients. These data imply that follow-up studies analyzing BM function during long-term G-CSF stimulation are required.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Adulto , Progresión de la Enfermedad , Eritrocitos/efectos de los fármacos , Femenino , Granulocitos/efectos de los fármacos , Humanos , Recuento de Leucocitos , Macrófagos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Fármacos Neuroprotectores/efectos adversos , Fármacos Neuroprotectores/uso terapéutico , Recuento de Plaquetas , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico
8.
J Thromb Haemost ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38936699

RESUMEN

BACKGROUND: Acquired hemophilia A (AHA) is an autoimmune bleeding disorder caused by neutralizing antibodies against coagulation factor VIII (FVIII). Immunosuppressive therapy (IST) is standard of care to eradicate autoantibody production and protect from further bleeding but carries a risk of severe infection and mortality in frail patients with AHA. Recently, emicizumab has been studied for its potential to reduce the need for early and aggressive IST. OBJECTIVE: To compare outcomes of two studies that used either IST (GTH-AH 01/2010; n=101) or prophylaxis with emicizumab (GHT-AHA-EMI; n=47) early after diagnosis of AHA. METHODS: Baseline characteristics were balanced by propensity score matching. Primary endpoint was the rate of clinically relevant new bleeds during the first 12 weeks; secondary endpoints were adverse events and overall survival. RESULTS: The negative binominal model-based bleeding rate was 68% lower with emicizumab as compared with IST (incident rate ratio 0.325, 95% confidence interval (CI) 0.182-0.581). No difference was apparent in the overall frequency of infections (emicizumab 21%, IST 29%) during the first 12 weeks, but infections were less often fatal in emicizumab treated patients (0%) compared with IST treated patients (11%). Thromboembolic events occurred less often with emicizumab (2%) than IST (7%). Overall survival after 24 weeks was better with emicizumab (90% versus 76%, HR 0.44; 95% CI 0.24-0.81). CONCLUSION: Using emicizumab instead of IST in the early phase after initial diagnosis of AHA reduced bleeding and fatal infections, and improved overall survival.

9.
Sci Rep ; 13(1): 6154, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-37061580

RESUMEN

Lipids play a central role in platelet physiology. Changes in the lipidome have already been described for basal and activated platelets. However, quantitative lipidomic data of platelet activation, including the released complex lipids, are unavailable. Here we describe an easy-to-use protocol based on flow-injection mass spectrometry for the quantitative analysis of bulk lipid species in basal and activated human platelets and their lipid release after thrombin activation. We provide lipid species concentrations of 12 healthy human donors, including cholesteryl ester (CE), ceramide (Cer), free cholesterol (FC), hexosylceramide (HexCer), lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), phosphatidylcholine (PC), phosphatidylethanolamine (PE), phosphatidylinositol (PI), phosphatidylserine (PS), sphingomyelin (SM) and triglycerides (TG). The assay exhibited good technical repeatability (CVs < 5% for major lipid species in platelets). Except for CE and TG, the inter-donor variability of the majority of lipid species concentrations in platelets was < 30% CV. Balancing of concentrations revealed the generation of LPC and loss of TG. Changes in lipid species concentrations indicate phospholipase-mediated release of arachidonic acid mainly from PC, PI, and PE but not from PS. Thrombin induced lipid release was mainly composed of FC, PS, PC, LPC, CE, and TG. The similarity of the released lipidome with that of plasma implicates that lipid release may originate from the open-canalicular system (OCS). The repository of lipid species concentrations determined with this standardized platelet release assay contribute to elucidating the physiological role of platelet lipids and provide a basis for investigating the platelet lipidome in patients with hemorrhagic or thrombotic disorders.


Asunto(s)
Plaquetas , Trombina , Humanos , Espectrometría de Masas , Triglicéridos , Colesterol , Lecitinas
10.
Blood Adv ; 7(1): 122-130, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947142

RESUMEN

The root cause of autoantibody formation against factor VIII (FVIII) in acquired hemophilia A (AHA) remains unclear. We aimed to assess whether AHA is exclusively associated with autoantibodies toward FVIII or whether patients also produce increased levels of autoantibodies against other targets. A case-control study was performed enrolling patients with AHA and age-matched controls. Human epithelial cell (HEp-2) immunofluorescence was applied to screen for antinuclear (ANA) and anticytoplasmic autoantibodies. Screening for autoantibodies against extractable nuclear antigens was performed by enzyme immunoassay detecting SS-A/Ro, SS-B/La, U1RNP, Scl-70, Jo-1, centromere B, Sm, double-stranded DNA, and α-fodrin (AF). Patients with AHA were more often positive for ANA than control patients (64% vs 30%; odds ratio [OR] 4.02, 1.98-8.18) and had higher ANA titers detected than controls. Cytoplasmic autoantibodies and anti-AF immunoglobulin A autoantibodies were also more frequent in patients with AHA compared with controls. Autoantibodies against any target other than FVIII were found in 78% of patients with AHA compared with 46% of controls (OR 4.16, 1.98-8.39). Results were similar preforming sensitivity analyses (excluding either subjects with autoimmune disorders, cancer, pregnancy, or immunosuppressive medication at baseline) and in multivariable binary logistic regression. To exclude that autoantibody staining was merely a result of cross-reactivity of anti-FVIII autoantibodies, we tested a mix of 7 well-characterized monoclonal anti-FVIII antibodies. These antibodies did not stain HEp-2 cells used for ANA detection. In conclusion, a diverse pattern of autoantibodies is associated with AHA, suggesting that a more general breakdown of immune tolerance might be involved in its pathology.


Asunto(s)
Hemofilia A , Hemostáticos , Humanos , Autoanticuerpos , Estudios de Casos y Controles , Factor VIII , Hemofilia A/diagnóstico
11.
Hamostaseologie ; 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-38049124

RESUMEN

BACKGROUND: Acquired hemophilia A (AHA) is a severe bleeding disorder caused by autoantibodies against coagulation factor VIII (FVIII). Standard treatment consists of bleeding control with bypassing agents and immunosuppressive therapy. Emicizumab is a bispecific antibody that mimics the function of activated FVIII irrespective of the presence of neutralizing antibodies. Recently, the GTH-AHA-EMI study demonstrated that emicizumab prevents bleeds and allows to postpone immunosuppression, which may influence future treatment strategies. AIM: To provide clinical practice recommendations on the use of emicizumab in AHA. METHODS: A Delphi procedure was conducted among 33 experts from 16 German and Austrian hemophilia care centers. Statements were scored on a scale of 1 to 9, and agreement was defined as a score of ≥7. Consensus was defined as ≥75% agreement among participants, and strong consensus as ≥95% agreement. RESULTS: Strong consensus was reached that emicizumab is effective for bleed prophylaxis and should be considered from the time of diagnosis (100% consensus). A fast-loading regimen of 6 mg/kg on day 1 and 3 mg/kg on day 2 should be used if rapid bleeding prophylaxis is required (94%). Maintenance doses of 1.5 mg/kg once weekly should be given (91%). Immunosuppression should be offered to patients on emicizumab if they are eligible based on physical status (97%). Emicizumab should be discontinued when remission of AHA is achieved (97%). CONCLUSION: These GTH consensus recommendations provide guidance to physicians on the use of emicizumab in AHA and follow the results of clinical trials that have shown emicizumab is effective in preventing bleeding in AHA.

12.
Lancet Haematol ; 10(11): e913-e921, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37858328

RESUMEN

BACKGROUND: Acquired haemophilia A is caused by neutralising autoantibodies against coagulation factor VIII, leading to severe bleeding. Standard treatment involves immunosuppressive therapy, which is associated with adverse events and mortality in the frail population of patients with acquired haemophilia A. This study investigated whether emicizumab, a factor VIIIa mimetic antibody, protects patients with acquired haemophilia A from bleeding and allows deferral of immunosuppression during the first 12 weeks after diagnosis. METHODS: We report final results of an open-label, single-arm, phase 2 clinical trial. Adult patients with acquired haemophilia A from 16 haemophilia treatment centres in Germany and Austria were eligible if they had not previously received immunosuppression. Patients received emicizumab subcutaneously (6 and 3 mg/kg on days 1 and 2, 1·5 mg/kg weekly until week 12), but no immunosuppression. Follow-up was until week 24. The primary endpoint was the number of clinically relevant bleeds per patient-week until week 12. Emicizumab was considered effective if the mean bleeding rate was significantly below 0·15 bleeds per patient-week, the rate observed in a previous study of patients with acquired haemophilia A treated with bypassing agents and immunosuppression but no emicizumab. The study is registered with clinicaltrials.gov, NCT04188639 and is complete. FINDINGS: Of 49 patients screened from March 25, 2021, to June 10, 2022, 47 were enrolled (23 women, 24 men). Median age was 76 years (IQR 66-80), 46 (98%) of 47 patients were White, median factor VIII activity was 1·4 IU/dL (0·3-5·6), and median inhibitor concentration was 11·4 Bethesda units per mL (3·9-42·7). Mean breakthrough bleeding rate was 0·04 bleeds per patient-week (upper 97·5% CI 0·06). 33 (70%) of 47 patients had no bleeding events, seven patients (15%) had one bleed, six patients (13%) had two bleeds, and one patient (2%) had three bleeds. Adverse events of grade 3 or worse included COVID-19 (n=2), acute kidney injury (n=2), and stroke (n=1). Four of 47 patients died, including two deaths related to bleeding, one from COVID-19, and one from cardiac arrest (none were judged as related to emicizumab). INTERPRETATION: This study suggests that emicizumab prophylaxis prevents bleeding in patients with acquired haemophilia A and that immunosuppressive therapy can be deferred while patients are receiving this treatment. The low number of thromboembolic events, severe infections, and fatalities observed in this study are promising. FUNDING: This study was supported by funding from Hoffman-La Roche.


Asunto(s)
COVID-19 , Hemofilia A , Masculino , Adulto , Humanos , Femenino , Anciano , Hemofilia A/tratamiento farmacológico , Factor VIII/uso terapéutico , Hemorragia/etiología , Hemorragia/prevención & control , Hemorragia/tratamiento farmacológico
13.
Soc Psychiatry Psychiatr Epidemiol ; 47(10): 1581-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22202973

RESUMEN

PURPOSE: To establish the prevalence of victimisation in a UK population-based sample and to investigate the association between mental disorder and victimisation in both cross-sectional and prospective manner, whilst adjusting for potential confounds. METHODS: Data from the National Child Development Study (NCDS) were used to examine criminal victimisation, violent victimisation, and mental disorder at age 46 yerars, and also to measure history of mental disorder, when cohort members were aged 23, 33 and 42 years. Variables considered to be potential confounders or mediators of the association, including socio-economic status, family income, financial strain, education, housing ownership status, heavy drinking and gender, all measured at age 46 years, were considered in multivariate analyses. RESULTS: The prevalence of criminal victimisation amongst cohort members in the 12 months preceding interview was 15%; 2.2% of the participants reported experiencing violent victimisation in the past year. Mental disorder at age 46 was significantly associated with criminal and violent victimisation, even after adjusting for potential confounds. A prior history of mental disorder was found to be a robust predictor of criminal and violent victimisation. CONCLUSIONS: This study provides evidence that those with a mental disorder are at elevated risk of victimisation, including violent victimisation. That such an association might reflect an underlying causal relationship is further supported by the confirmation that the association holds true when mental disorder is measured well before the assessment of victimisation risk, and that it persists despite adjustment for a number of potential confounding factors.


Asunto(s)
Víctimas de Crimen/psicología , Trastornos Mentales/psicología , Violencia/psicología , Adulto , Trastornos Relacionados con Alcohol/complicaciones , Trastornos Relacionados con Alcohol/epidemiología , Víctimas de Crimen/estadística & datos numéricos , Estudios Transversales , Femenino , Humanos , Entrevistas como Asunto , Masculino , Trastornos Mentales/epidemiología , Persona de Mediana Edad , Análisis Multivariante , Vigilancia de la Población , Prevalencia , Estudios Prospectivos , Factores de Riesgo , Distribución por Sexo , Factores Socioeconómicos , Reino Unido/epidemiología , Violencia/estadística & datos numéricos , Poblaciones Vulnerables , Adulto Joven
14.
Hamostaseologie ; 42(5): 285-286, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36343647

RESUMEN

In the last couple of years sex specific health issues have continually been gaining attraction by physicians of different medical specialities. Sex differences have been described e.g. in the pathogenesis and mortality in patients affected by COVID-19, in metabolic regulation and cancer mechanisms.1 2 3 In the field of haemostasis, many aspects concerning risk factors, clinical presentation and management of thromboembolic disease and bleeding disorders also display sex differences.


Asunto(s)
Trastornos de la Coagulación Sanguínea , COVID-19 , Trombosis , Masculino , Femenino , Humanos , Hemostasis , Salud de la Mujer
15.
Hamostaseologie ; 42(5): 290-299, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36323276

RESUMEN

The lifetime risk of venous thromboembolism (VTE) is slightly higher in women than in men. There are several issues related to VTE that are unique to women. Combined hormonal contraceptives and pregnancy increase the risk of VTE in women of childbearing age, whereas hormone replacement therapy increases the VTE risk of postmenopausal women. Hereditary thrombophilia and risk factors such as older age, obesity, or smoking contribute to the risk increase. In women diagnosed with acute hormone-related VTE who are treated with oral anticoagulants, adequate contraception is mandatory to avoid unwanted pregnancies. According to current knowledge, hormonal contraception may be continued during anticoagulant therapy but must be switched to an estrogen-free contraception method at least 6 weeks before the termination of anticoagulation. VTE is also a major cause of maternal morbidity and mortality during pregnancy and the postpartum period. Currently, assisted reproduction technologies such as in vitro fertilization are widely used to treat couples affected by infertility. Complications of fertility treatment comprise VTE cases, especially in women with ovarian hyperstimulation syndrome. With this review, we intended to focus on VTE issues in women and summarize current evidence and guideline recommendations.


Asunto(s)
Trombofilia , Tromboembolia Venosa , Embarazo , Masculino , Femenino , Humanos , Tromboembolia Venosa/etiología , Tromboembolia Venosa/complicaciones , Trombofilia/complicaciones , Anticoagulantes/efectos adversos , Factores de Riesgo , Anticoncepción
16.
Hamostaseologie ; 42(5): 320-329, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36323279

RESUMEN

Antithrombin (AT) deficiency is a high-risk thrombophilia and a rare condition. The risk of venous thromboembolism (VTE) is increased in AT-deficient women during pregnancy and the postpartum period and is especially high in women with a prior history of VTE. A thorough assessment of VTE risk is recommended in pregnant AT-deficient women, comprising the degree and type of AT deficiency, genetic mutations, personal and family history, and additional preexisting or pregnancy-specific risk factors. Due to a lack of adequate study data, there is limited guidance on the management of AT deficiency in pregnancy, including the need for prophylactic anticoagulation, the appropriate dose of low-molecular-weight heparin (LMWH), and the role of AT substitution. LMWH is the medication of choice for the pharmacological prophylaxis and treatment of VTE in pregnancy. Patients with a history of VTE should receive full-dose LMWH during pregnancy and the postpartum period. AT concentrates are a treatment option when anticoagulation is withheld in potentially high-risk events such as childbirth, bleeding, or surgery and in cases of acute VTE despite the use of therapeutic dose anticoagulation. Women with AT deficiency should be counseled at specialized centers for coagulation disorders or vascular medicine, and close cooperation between obstetricians and anesthesiologists is warranted before delivery and during the peripartum period.


Asunto(s)
Deficiencia de Antitrombina III , Tromboembolia Venosa , Embarazo , Humanos , Femenino , Heparina de Bajo-Peso-Molecular/uso terapéutico , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/prevención & control , Deficiencia de Antitrombina III/complicaciones , Deficiencia de Antitrombina III/diagnóstico , Deficiencia de Antitrombina III/tratamiento farmacológico , Anticoagulantes/uso terapéutico , Antitrombinas/uso terapéutico , Factores de Riesgo
17.
Clin Res Hepatol Gastroenterol ; 46(8): 101932, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35504460

RESUMEN

Despite one of the largest vaccination campaigns in human history, the COVID-19 pandemic has not been yet defeated. More than 10 billion doses of COVID-19 vaccine have been administered worldwide. AstraZeneca's Vaxzevria (ChAdOx1 nCoV-19 / AZD1222) was approved as the first viral vector-based vaccine in the EU on 29 January 2021. Thromboembolic events are a rare complication of vaccination with ChAdOx1 nCoV-19 in the context of, now known as vaccine-induced immune thrombotic thrombocytopenia (VITT), with an incidence of 1.5-3 in 100,000 vaccinations. VITT is clinically as well as pathophysiologically comparable to heparin-induced thrombocytopenia. Illustrated by a fulminant patient case, a multidisciplinary step-by-step guideline was developed for the recognition, diagnosis, and management of patients with severe acute portosplanchic venous thrombosis with mesenteric ischemia due to vaccine-induced immunogenic thrombotic thrombocytopenia.


Asunto(s)
COVID-19 , Hepatopatías , Trombocitopenia , Trombosis , Trombosis de la Vena , Vacunas contra la COVID-19/efectos adversos , ChAdOx1 nCoV-19 , Humanos , Hepatopatías/complicaciones , Pandemias , Vena Porta , Trombocitopenia/inducido químicamente , Trombocitopenia/complicaciones , Trombosis/complicaciones , Vacunación/efectos adversos , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/etiología
18.
PLoS One ; 17(8): e0272577, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35939484

RESUMEN

OBJECTIVES: Unfractionated heparin (UFH) is the commonly used anticoagulant to prevent clotting of the ECMO circuit and thrombosis of the cannulated vessels. A side effect of UFH is heparin-induced thrombocytopenia (HIT). Little is known about HIT during ECMO and the impact of changing anticoagulation in ECMO patients with newly diagnosed HIT. The aim of the study was to determine the prevalence, complications, impact of switching anticoagulation to argatroban and outcomes of patients developing heparin-induced thrombocytopenia (HIT) during either veno-venous (VV) or veno-arterial (VA) ECMO. METHODS: Retrospective observational single centre study of prospectively collected data of consecutive patients receiving VV ECMO therapy for severe respiratory failure and VA ECMO for circulatory failure from January 2006 to December 2016 of the Medical intensive care unit (ICU) of the University Hospital of Regensburg. Treatment of HIT on ECMO was done with argatroban. RESULTS: 507 patients requiring ECMO were included. Further HIT-diagnostic was conducted if HIT-4T-score was ≥4. The HIT-confirmed group had positive HIT-enzyme-linked-immunosorbent-assay (ELISA) and positive heparin-induced-platelet-activation (HIPA) test, the HIT-suspicion group a positive HIT-ELISA and missing HIPA but remained on alternative anticoagulation until discharge and the HIT-excluded group a negative or positive HIT-ELISA, however negative HIPA. These were compared to group ECMO-control without any HIT suspicion. The prevalence of HIT-confirmed was 3.2%, of HIT-suspicion 2.0% and HIT-excluded 10.8%. Confirmed HIT was trendwise more frequent in VV than in VA (3.9 vs. 1.7% p = 0.173). Compared to the ECMO control group, patients with confirmed HIT were longer on ECMO (median 13 vs. 8 days, p = 0.002). Different types of complications were higher in the HIT-confirmed than in the ECMO-control group, but in-hospital mortality was not different (31% vs. 41%, p = 0.804). CONCLUSION: HIT is rare on ECMO, should be suspected, if platelets are decreasing, but seems not to increase mortality if treated promptly.


Asunto(s)
Oxigenación por Membrana Extracorpórea , Trombocitopenia , Anticoagulantes/efectos adversos , Oxigenación por Membrana Extracorpórea/efectos adversos , Heparina/efectos adversos , Humanos , Prevalencia , Estudios Retrospectivos , Trombocitopenia/inducido químicamente , Trombocitopenia/epidemiología , Trombocitopenia/terapia
19.
Dtsch Med Wochenschr ; 146(11): 705-709, 2021 06.
Artículo en Alemán | MEDLINE | ID: mdl-34062582

RESUMEN

The use of combined hormonal contraceptives (CHC) is a well-established risk factor for venous thromboembolism (VTE). The VTE risk depends on the specific combination of oestrogen and gestagen components. Progestin-only contraceptives with the exception of depot medroxyprogesterone acetate are not associated with a significant VTE risk and can therefore be offered to women with known thrombophilia or a prior VTE. The recent German S3 guideline "Contraception" advises to carefully assess individual VTE risk factors before prescribing CHC. According to recent data there is no evidence suggesting that VTE risk is increased during oral anticoagulation. To reduce the risk of vaginal bleeding complication and the risk of unplanned pregnancy, the use of CHC can be continued under anticoagulation treatment provided that the patient is switched to an oestrogen-free contraception no later than six weeks before the end of anticoagulation. The risk of recurrence is low in women with hormone-associated VTE. Anticoagulation is therefore in general discontinued after 3-6 months. Thromboprophylaxis with low molecular heparin is recommended for women with prior hormone-associated VTE during pregnancy and the postpartum period.


Asunto(s)
Anticoncepción Hormonal/efectos adversos , Tromboembolia Venosa , Anticoagulantes/uso terapéutico , Femenino , Humanos , Embarazo , Tromboembolia Venosa/inducido químicamente , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/prevención & control
20.
Hematology ; 26(1): 565-576, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34384344

RESUMEN

Objectives: Multipotent mesenchymal stromal cells (MSC) play a pivotal role in the bone marrow (BM) niche. Stanniocalcin 1 (STC1) secreted by MSC has been demonstrated to promote the survival of neoplastic cells and was suggested a marker for minimal residual disease of acute myeloid leukemia (AML). Therefore, we evaluated the expression of STC1 in MSC from AML patients (MSCAML) compared to MSC from healthy donors (MSCHD).Methods: Liquid culture assays of MSCAML and MSCHD were performed to compare expansion capacity. Gene expression profiles of MSCAML vs. MSCHD were established. Secretion of STC1 was tested by ELISA in MSCAML vs. MSCHD and expression of STC1 in AML- vs. HD-BM by immunohistochemistry. In addition, co-cultures of AML cells on MSC were initiated and ultrastructural intercellular communication patterns were investigated. Finally, the effect of blocking STC1 on AML cells was evaluated.Results: MSCAML showed significant decreased expansion capacity compared to MSCHD. Gene analysis revealed marked overexpression of STC1 in MSCAML. ELISA and immunohistochemical findings confirmed this observation. Electron microscopy analysis showed reciprocal stimulation between AML cells and MSC. Blockade of STC1 did not significantly affect AML cell proliferation and apoptosis.Discussion: Characteristics of MSC differ depending on whether they originate from AML patients or from HD. STC1 was mostly overexpressed in MSCAML compared to MSCHD. In vitro blockade of STC1, however, was not associated with AML cell proliferation and apoptosis.Conclusion: Differences in expression levels of glycoproteins from MSCAML compared to MSCHD not necessarily assume that these molecules are niche-relevant in leukemic disease.


Asunto(s)
Glicoproteínas/genética , Leucemia Mieloide Aguda/genética , Células Madre Mesenquimatosas/patología , Regulación hacia Arriba , Adulto , Anciano , Células Cultivadas , Femenino , Glicoproteínas/análisis , Humanos , Leucemia Mieloide Aguda/patología , Masculino , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA