Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
Int J Mol Sci ; 24(3)2023 Jan 23.
Article in English | MEDLINE | ID: covidwho-2253589

ABSTRACT

Splanchnic vein thrombosis is a rare but potentially life-threatening manifestation of venous thromboembolism, with challenging implications both at the pathological and therapeutic level. It is frequently associated with liver cirrhosis, but it could also be provoked by myeloproliferative disorders, cancer of various gastroenterological origin, abdominal infections and thrombophilia. A portion of splanchnic vein thrombosis is still classified as idiopathic. Here, we review the mechanisms of splanchnic vein thrombosis, including new insights on the role of clonal hematopoiesis in idiopathic SVT pathogenesis, with important implications from the therapeutic standpoint.


Subject(s)
Myeloproliferative Disorders , Venous Thromboembolism , Venous Thrombosis , Humans , Venous Thrombosis/complications , Myeloproliferative Disorders/complications
3.
Cancer Rep (Hoboken) ; 6(1): e1769, 2023 01.
Article in English | MEDLINE | ID: covidwho-2238549

ABSTRACT

BACKGROUND: T-lymphoblastic lymphoma (T-LBL) is an aggressive malignancy of T-lymphoid precursors, rarely co-occurring with myeloid/lymphoid neoplasms with eosinophilia (M/LNs-Eo), with consequent rearrangement of tyrosine kinase (TK)-related genes. The FIP1L1-PDGFRA fusion gene is the most frequent molecular abnormality seen in eosinophilia-associated myeloproliferative disorders, but is also present in acute myeloid leukemia (AML), T-lymphoblastic leukemia/lymphoma (TLL), or both simultaneously. T-LBL mainly affects children and young adults, involving lymph node, bone marrow, and thymus. It represents about 85% of all immature lymphoblastic lymphomas, whereas immature B-cell lymphomas comprise approximately 15% of all cases of LBL. CASE: In this case report, we present an example of T cell lymphoblastic lymphoma with coexistent eosinophelia, treated successfully with a tyrosine-kinase inhibitor (TKI). CONCLUSION: FIP1L1-PDGFRA-positive T-LBL and myeloproliferative disorders have excellent response to low-dose treatment with (TKI) imatinib. Most patients achieve rapid and complete hematologic and molecular remission within weeks.


Subject(s)
Eosinophilia , Myeloproliferative Disorders , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor alpha/therapeutic use , Imatinib Mesylate/therapeutic use , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Eosinophilia/diagnosis , Eosinophilia/drug therapy , Eosinophilia/genetics , Protein Kinase Inhibitors/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Oncogene Proteins, Fusion/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/therapeutic use
5.
Blood Adv ; 6(22): 5857-5865, 2022 11 22.
Article in English | MEDLINE | ID: covidwho-2043101

ABSTRACT

IDH2 (isocitrate dehydrogenase 2) mutations occur in approximately 15% of patients with acute myeloid leukemia (AML). The IDH2 inhibitor enasidenib was recently approved for IDH2-mutated relapsed or refractory AML. We conducted a multi-center, phase I trial of maintenance enasidenib following allogeneic hematopoietic cell transplantation (HCT) in patients with IDH2-mutated myeloid malignancies. Two dose levels, 50mg and 100mg daily were studied in a 3 × 3 dose-escalation design, with 10 additional patients treated at the recommended phase 2 dose (RP2D). Enasidenib was initiated between days 30 and 90 following HCT and continued for twelve 28-day cycles. Twenty-three patients were enrolled, of whom 19 initiated post-HCT maintenance. Two had myelodysplastic syndrome, and 17 had AML. All but 3 were in first complete remission. No dose limiting toxicities were observed, and the RP2D was established at 100mg daily. Attributable grade ≥3 toxicities were rare, with the most common being cytopenias. Eight patients stopped maintenance before completing 12 cycles, due to adverse events (n=3), pursuing treatment for graft-vs-host disease (GVHD) (n=2), clinician choice (n=1), relapse (n=1), and COVID infection (n=1). No cases of grade ≥3 acute GVHD were seen, and 12-month cumulative incidence of moderate/severe chronic GVHD was 42% (20-63%). Cumulative incidence of relapse was 16% (95% CI: 3.7-36%); 1 subject relapsed while receiving maintenance. Two-year progression-free and overall survival were 69% (95% CI: 39-86%) and 74% (95% CI, 44-90%), respectively. Enasidenib is safe, well-tolerated, with preliminary activity as maintenance therapy following HCT, and merits additional study. The study was registered at www.clinicaltrials.gov (#NCT03515512).


Subject(s)
COVID-19 , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Recurrence
7.
Oncology ; 100(8): 460-466, 2022.
Article in English | MEDLINE | ID: covidwho-1962525

ABSTRACT

INTRODUCTION: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral agent responsible for the coronavirus disease of 2019. The disease is primarily a respiratory illness; however, multisystem involvement is not uncommon. The infection is reported to be more severe in patients with multiple comorbidities and immunocompromised patients. Patients with hematological malignancies are immunocompromised and prone to develop severe SARS-CoV-2 infection. The SARS-CoV-2 had developed several mutations that resulted in different strains with different virulence and different degree of protection by vaccination or prior infection. The Omicron variant is reported to cause mild illness; however, the effect on patients with hematological malignancies like myeloproliferative neoplasms (MPNs) is not clear. We present patients with MPNs who had infection with the Omicron variant of the SARS-CoV-2 and their outcomes. METHODS: Retrospective data from the National Center for Cancer Care and Research records from December 20, 2021, to January 30, 2022. Participants were adults over the age of 18 years with Omicron infection who had been diagnosed with Philadelphia-negative MPNs, essential thrombocythemia, polycythemia vera (PV), and primary myelofibrosis according to the 2008/2016 WHO classification for MPN. RESULTS: Twenty-two patients with Philadelphia-negative MPN had Omicron infection. All patients had a mild disease according to the WHO classification of COVID-19 severity. Most of the patients had medical comorbidities, with hypertension being the most common comorbidity. However, only one patient with PV required hospitalization. DISCUSSION/CONCLUSIONS: In patients with Philadelphia-negative MPN, the Omicron variant of SARS-CoV-2 usually results in mild infection.


Subject(s)
COVID-19 , Myeloproliferative Disorders , Polycythemia Vera , Adult , Humans , Middle Aged , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Retrospective Studies , SARS-CoV-2
8.
Rinsho Ketsueki ; 63(6): 647-654, 2022.
Article in Japanese | MEDLINE | ID: covidwho-1934597

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has made the management of coronavirus disease-2019 (COVID-19) in patients with hematological disorders a new and important theme for hematologists. Patients with myeloproliferative neoplasms (MPNs) are susceptible to SARS-CoV-2 and are at an increased risk of death after the onset of COVID-19. Thus, infection prevention measures, including vaccination for all patients, are important. Patients with MPNs who have COVID-19 have a poor prognosis, as do patients with other hematological malignancies. The thrombogenic characteristics of MPNs increase the risk of venous thrombosis due to COVID-19. Anticoagulant therapy is adjusted according to the risk of each case after COVID-19 onset. However, thrombosis occurs at a high rate, especially in patients with essential thrombocythemia. Additionally, patients with myelofibrosis have an increased risk of death and bleeding. Ruxolitinib treatment poses a risk of SARAS-CoV-2 infection, and its abrupt discontinuation after infection is associated with an increased risk of death. The emerging evidence of COVID-19 has been quickly reflected in the available treatment recommendations and guidelines.


Subject(s)
COVID-19 , Hematologic Neoplasms , Myeloproliferative Disorders , Thrombocythemia, Essential , COVID-19/complications , Hematologic Neoplasms/complications , Humans , Myeloproliferative Disorders/complications , SARS-CoV-2 , Thrombocythemia, Essential/complications
9.
Ann Hematol ; 101(8): 1825-1834, 2022 Aug.
Article in English | MEDLINE | ID: covidwho-1858973

ABSTRACT

COVID-19 in patients with hematological diseases is associated with a high mortality. Moreover, preventive vaccination demonstrated reduced efficacy and the knowledge on influencing factors is limited. In this single-center study, antibody levels of the SARS-CoV-2 spike protein were measured ≥ 2 weeks after 2nd COVID-19 vaccination with a concentration ≥ 0.8 U/mL considered positive. Between July and October 2021, in a total of 373 patients (median age 64 years, 44% women) with myeloid neoplasms (n = 214, 57%), lymphoid neoplasms (n = 124, n = 33%), and other diseases (n = 35, 10%), vaccination was performed with BNT162b2 (BioNTech), mRNA-1273 (Moderna), ChADOx1 (AstraZeneca), or a combination. A total of 229 patients (61%) were on active therapy within 3 months prior vaccination and 144 patients (39%) were previously treated or treatment naïve. Vaccination-related antibody response was negative in 56/373 patients (15%): in 39/124 patients with lymphoid neoplasms, 13/214 with myeloid neoplasms, and 4/35 with other diseases. Active treatment per se was not correlated with negative response. However, rituximab and BTK inhibitor treatment were correlated significantly with a negative vaccination response, whereas younger age and chronic myeloid leukemia (CML) disease were associated with positive response. In addition, 5 of 6 patients with myeloproliferative neoplasm (MPN) and negative vaccination response were on active treatment with ruxolitinib. In conclusion, a remarkable percentage of patients with hematological diseases had no response after 2nd COVID-19 vaccination. Multivariable analysis revealed important factors associated with response to vaccination. The results may serve as a guide for better protection and surveillance in this vulnerable patient cohort.


Subject(s)
Antibody Formation , COVID-19 Vaccines , COVID-19 , Hematologic Diseases , Antibodies, Viral , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Female , Hematologic Diseases/complications , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Male , Middle Aged , Myeloproliferative Disorders/complications , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Treatment Failure , Vaccination
11.
12.
Br J Haematol ; 197(6): 691-696, 2022 06.
Article in English | MEDLINE | ID: covidwho-1714140

ABSTRACT

Data on the response to the COVID-19 vaccine in patients with myeloid malignancy, who are at severe risk in case of infection, have not emerged. In a study of 69 patients with myeloid malignancies, including 46 patients with acute myeloid leukaemia (AML) and 23 patients with myelodysplastic syndrome (MDS), anti-spike SARS-CoV-2 antibody titres were measured 3 months after the second mRNA-based vaccination. Seroconversion rates for AML and MDS were 94.7% and 100% respectively, with no significant difference from healthy controls (HCs). Patients with MDS showed a significantly lower antibody titre than that in HCs or AML patients. In AML patients, the antibody titres were comparable to those in HCs when treatment was completed, but lower in patients under maintenance therapy. The response to COVID-19 vaccine appears to be related to disease and treatment status. Patients with myeloid malignancies may be more responsive to vaccines than patients with lymphoid malignancies.


Subject(s)
COVID-19 , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Myeloproliferative Disorders , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , RNA, Messenger , SARS-CoV-2 , Vaccination
14.
Hematology Am Soc Hematol Educ Program ; 2021(1): 710-717, 2021 12 10.
Article in English | MEDLINE | ID: covidwho-1566502

ABSTRACT

Both myeloproliferative neoplasms (MPNs) and coronavirus disease 2019 (COVID-19) are characterized by an intrinsic thrombotic risk. Little is known about the incidence and the outcome of thrombotic events in patients with MPN infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but common mechanisms of coagulation activation, typical of both disorders, suggest that these patients can be at particularly high risk. To define the best thromboprophylaxis and treatment regimens in both MPN and COVID-19, individual- and disease-specific thrombotic risk factors, bleeding risk, and concomitant specific treatments need to be considered. In this case-based review, an individualized approach is presented in a case of SARS-CoV-2 infection occurring in a man with polycythemia vera (PV). A primary anticoagulant thromboprophylaxis strategy and adjustment of his PV treatment were implemented. However, during the hospital stay, he experienced pulmonary embolism and therapeutic anticoagulation had to be set. Then his condition improved, and discharge was planned. Postdischarge decisions had to be made about the type and duration of venous thromboembolism treatment as well as the management of PV-specific drugs. The steps of our decisions and recommendations are presented.


Subject(s)
COVID-19/complications , Myeloproliferative Disorders/complications , Polycythemia Vera/complications , Thrombosis/drug therapy , Thrombosis/etiology , Aged , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , COVID-19/blood , Humans , Male , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/drug therapy , Polycythemia Vera/blood , Polycythemia Vera/drug therapy , Risk Factors , Thrombosis/blood , COVID-19 Drug Treatment
17.
researchsquare; 2021.
Preprint in English | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-1053230.v1

ABSTRACT

Background: Patients with hematologic neoplasm may have compromised immunity due to their malignancy and/or treatment, and may be at elevated risk of severe COVID-19. However, the studies bring together patients with hematologic neoplasms and solid tumors into a single group, making no distinction about the types of hematological tumors and their treatments. This systematic review is designed to explore the risk of severe COVID-19 in patients with hematologic neoplasm. Studies about patients, adult or children, with hematologic neoplasm and COVID-19 will be included. Methods: : A systematic review according to Joanna Briggs Institute methodology for systematic reviews of etiology and risk will be performed. The review will consider as participants adults or children with COVID-19 infection detected by RT-PCR or serology (SARS-CoV-2 antibody). We will be included studies without routine labs confirmation of COVID-19 if the patients presented clinical/physical exam and computed tomography suggesting COVID-19. The exposure of interest will be hematologic neoplasm, which include lymphomas, acute and chronic leukemias, myeloma, myelodysplastic syndrome, and myeloproliferative diseases. We will consider cohort, case-control, analytical cross-sectional studies. Outcomes among patients with COVID-19 are critical symptoms, hospitalizations, intensive care unit admissions, mechanical ventilation and deaths. We will exclude studies with other neoplasms than hematologic neoplasms. Search strategies have been created for the Embase, Medline and LILACS. Two reviewers independently will assess the studies for their eligibility, will extract data and will evaluate their risk of bias. Similar outcomes measured in at least two studies will be plotted in the meta-analysis using the Joanna Briggs Institute System for the Unified Management, Assessment and Review of Information. Discussion: This systematic review aims to evaluate if patients with hematologic neoplasm may be at elevated risk of severe COVID-19. This review will differ from the previous ones because we will include controlled studies and groups with only hematologic neoplasm, excluding other cancers. The main hypothesis of our research is that not all hematological cancer patients have high risk of severe COVID-19. Trial registration number: PROSPERO CRD42020199318.


Subject(s)
Myelodysplastic Syndromes , Leukemia , Lymphoma , Myeloproliferative Disorders , Neoplasms , Hematologic Neoplasms , COVID-19 , Multiple Myeloma
20.
Curr Hematol Malig Rep ; 16(5): 455-463, 2021 10.
Article in English | MEDLINE | ID: covidwho-1442179

ABSTRACT

PURPOSE OF REVIEW: Coronavirus disease 2019 (COVID-19) is associated with a high rate of respiratory failure, thromboembolism, bleeding, and death. Patients with myeloproliferative neoplasms (MPNs) are prone to both thrombosis and bleeding, calling for special care during COVID-19. We reviewed the clinical features of MPN patients with COVID-19, suggesting guidance for treatment. RECENT FINDINGS: One study by the European LeukemiaNet collected 175 MPN patients with COVID-19 during the first wave of the pandemic, from February to May 2020. Patients with primary myelofibrosis (PMF) were at higher risk of mortality (48%) in comparison with essential thrombocythemia (ET) (25%) and polycythemia vera (19%); the risk of death was higher in those patients who abruptly discontinued ruxolitinib. In patients followed at home, in regular wards, or in ICU, the thrombosis rate was 1.0%, 2.8%, and 18.4%, respectively. Independent risk factors for thrombosis were ET phenotype, transfer to ICU, and neutrophil/lymphocyte ratio; major bleeding occurred in 4.3% of patients, particularly those with PMF. MPN patients with non-severe COVID-19 treated at home should continue their primary or secondary antithrombotic prophylaxis with aspirin or oral anticoagulants. In the case of hospitalization, patients assuming aspirin should add low molecular weight heparin (LMWH) at standard doses. In contrast, LMWH at intermediate/therapeutic doses should replace oral anticoagulants prescribed for atrial fibrillation or previous venous thromboembolism. Intermediate/high doses of LMWH can also be considered in ICU patients with ET, particularly in the case of a rapid decline in the number of platelets and progressive respiratory failure.


Subject(s)
COVID-19 , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative , Anticoagulants/therapeutic use , COVID-19/complications , COVID-19/diagnosis , COVID-19/epidemiology , COVID-19/therapy , Hemorrhage/epidemiology , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/complications , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/diagnosis , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/epidemiology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/epidemiology , Myeloproliferative Disorders/therapy , Pandemics , SARS-CoV-2/physiology , Thrombosis/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL