Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Blood ; 140(11): 1200-1228, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35767897

RESUMO

The classification of myeloid neoplasms and acute leukemias was last updated in 2016 within a collaboration between the World Health Organization (WHO), the Society for Hematopathology, and the European Association for Haematopathology. This collaboration was primarily based on input from a clinical advisory committees (CACs) composed of pathologists, hematologists, oncologists, geneticists, and bioinformaticians from around the world. The recent advances in our understanding of the biology of hematologic malignancies, the experience with the use of the 2016 WHO classification in clinical practice, and the results of clinical trials have indicated the need for further revising and updating the classification. As a continuation of this CAC-based process, the authors, a group with expertise in the clinical, pathologic, and genetic aspects of these disorders, developed the International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias. Using a multiparameter approach, the main objective of the consensus process was the definition of real disease entities, including the introduction of new entities and refined criteria for existing diagnostic categories, based on accumulated data. The ICC is aimed at facilitating diagnosis and prognostication of these neoplasms, improving treatment of affected patients, and allowing the design of innovative clinical trials.


Assuntos
Neoplasias Hematológicas , Leucemia , Transtornos Mieloproliferativos , Doença Aguda , Consenso , Genômica , Neoplasias Hematológicas/patologia , Humanos , Leucemia/diagnóstico , Leucemia/genética , Leucemia/patologia , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Organização Mundial da Saúde
2.
Am J Hematol ; 99(5): 844-853, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38357714

RESUMO

Splenomegaly is the clinical hallmark of myelofibrosis. Splenomegaly at the time of allogeneic hematopoietic cell transplantation (HCT) is associated with graft failure and poor graft function. Strategies to reduce spleen size before HCT especially after failure to Janus kinase (JAK) inhibition represent unmet clinical needs in the field. Here, we leveraged a global collaboration to investigate the safety and efficacy of splenic irradiation as part of the HCT platform for patients with myelofibrosis. We included 59 patients, receiving irradiation within a median of 2 weeks (range, 0.9-12 weeks) before HCT. Overall, the median spleen size prior to irradiation was 23 cm (range, 14-35). Splenic irradiation resulted in a significant and rapid spleen size reduction in 97% of patients (57/59), with a median decrease of 5.0 cm (95% confidence interval, 4.1-6.3 cm). The most frequent adverse event was thrombocytopenia, with no correlation between irradiation dose and hematological toxicities. The 3-year overall survival was 62% (95% CI, 48%-76%) and 1-year non-relapse mortality was 26% (95% CI, 14%-38%). Independent predictors for survival were severe thrombocytopenia and anemia before irradiation, transplant-specific risk score, higher-intensity conditioning, and present portal vein thrombosis. When using a propensity score matching adjusted for common confounders, splenic irradiation was associated with significantly reduced relapse (p = .01), showing a 3-year incidence of 12% for splenic irradiation versus 29% for patients with immediate HCT and 38% for patients receiving splenectomy. In conclusion, splenic irradiation immediately before HCT is a reasonable approach in patients experiencing JAK inhibition failure and is associated with a low incidence of relapse.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Mielofibrose Primária , Trombocitopenia , Humanos , Baço , Esplenomegalia/etiologia , Esplenomegalia/radioterapia , Mielofibrose Primária/radioterapia , Mielofibrose Primária/complicações , Transplante de Células-Tronco Hematopoéticas/métodos , Trombocitopenia/complicações , Recidiva , Condicionamento Pré-Transplante/métodos , Doença Enxerto-Hospedeiro/etiologia
3.
J Thromb Thrombolysis ; 57(2): 186-193, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37839025

RESUMO

INTRODUCTION: Patients with myeloproliferative neoplasms (MPNs) and atrial fibrillation (AF) are at increased risk of thrombosis and bleeding. However, the risk of thrombosis and bleeding in patients with AF and MPN compared with the general population with AF is unclear. Additionally, traditional risk scores (CHA2DS2-VASC and HAS-BLED) for risk/benefit estimation of thromboprophylaxis in AF do not account for MPN status. Therefore, we aimed to investigate bleeding and thrombosis risk in patients with MPN hospitalized for AF. METHODS: We utilized the National Readmission Database (NRD) to identify patients with AF with and without MPN. Primary bleeding and thrombosis outcomes were in-hospital or 30-day readmission for bleeding or thrombosis, respectively. We propensity score (PS) matched patients with and without MPN. Risk of primary outcomes in MPN was assessed in PS matched cohort using logistic regression. Receiver operating characteristic (ROC) curve used to evaluate predictive ability of CHA2DS2-VASC and HAS-BLED of primary thrombosis and bleeding outcomes, respectively. RESULTS: 24,185 patients without MPN were matched with 1,617 patients with MPN and variables were balanced between groups. Patients with MPN were at increased risk of meeting the thrombosis (OR 1.98, 95% CI 1.23-3.21) but not bleeding (OR 0.87, 95% CI 0.63-1.19) primary outcomes. In MPN, CHA2DS2-VASC predicted thrombosis (C-statistic 0.66, 95% CI 0.54-0.78) but HAS-BLED was a poor predictor of bleeding (C-statistic 0.55, 95% CI 0.46-0.64). CONCLUSION: In patients with AF, MPN was associated with increased risk of bleeding and thrombosis. HAS-BLED scores did not accurately predict bleeding in MPN. Further investigation is needed to refine risk scores in MPN.


Assuntos
Fibrilação Atrial , Neoplasias , Acidente Vascular Cerebral , Trombose , Tromboembolia Venosa , Humanos , Fibrilação Atrial/tratamento farmacológico , Readmissão do Paciente , Anticoagulantes/uso terapêutico , Tromboembolia Venosa/tratamento farmacológico , Neoplasias/complicações , Hemorragia/induzido quimicamente , Fatores de Risco , Medição de Risco , Trombose/induzido quimicamente , Hospitais , Acidente Vascular Cerebral/etiologia
4.
Cancer ; 129(7): 1075-1084, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36655338

RESUMO

BACKGROUND: Patients with acute myeloid leukemia (AML) experience a substantial decline in quality of life (QoL) and mood during their hospitalization for intensive chemotherapy, yet few interventions have been developed to enhance patient-reported outcomes during treatment. METHODS: We conducted a pilot randomized trial (ClinicalTrials.gov identifier NCT03372291) of DREAMLAND, a psychological mobile application for patients with a new diagnosis of AML who are receiving intensive chemotherapy. Patients were randomly assigned to DREAMLAND or usual care. DREAMLAND included four required modules focused on: (1) supportive psychotherapy to help patients deal with the initial shock of diagnosis, (2) psychoeducation to manage illness expectations, (3) psychosocial skill-building to promote effective coping, and (4) self-care. The primary end point was feasibility, which was defined as ≥60% of eligible patients enrolling and 60% of those enrolled completing ≥60% of the required modules. We assessed patient QoL (the Functional Assessment of Cancer Therapy-Leukemia), psychological distress (the Hospital Anxiety and Depression Scale and the Patient Health Questionnaire-9), symptom burden (the Edmonton Symptom Assessment Scale), and self-efficacy (the Cancer Self-Efficacy Scale) at baseline and at day 20 after postchemotherapy. RESULTS: We enrolled 60 of 90 eligible patients (66.7%), and 62.1% completed ≥75% of the intervention modules. At day 20 after chemotherapy, patients who were randomized to DREAMLAND reported improved QoL scores (132.06 vs. 110.72; p =.001), lower anxiety symptoms (3.54 vs. 5.64; p = .010) and depression symptoms (Hospital Anxiety and Depression Scale: 4.76 vs. 6.29; p = .121; Patient Health Questionnaire-9: 4.62 vs. 8.35; p < .001), and improved symptom burden (24.89 vs. 40.60; p = .007) and self-efficacy (151.84 vs. 135.43; p = .004) compared with the usual care group. CONCLUSIONS: A psychological mobile application for patients with newly diagnosed AML is feasible to integrate during hospitalization for intensive chemotherapy and may improve QoL, mood, symptom burden, and self-efficacy.


Assuntos
Leucemia Mieloide Aguda , Aplicativos Móveis , Humanos , Qualidade de Vida/psicologia , Projetos Piloto , Ansiedade/terapia , Leucemia Mieloide Aguda/terapia , Depressão/psicologia
5.
J Oncol Pharm Pract ; 29(7): 1762-1765, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37282628

RESUMO

INTRODUCTION: Tyrosine kinase inhibitor (TKI) use leads to near-normal life expectancy in patients with chronic myeloid leukemia (CML); unfortunately for some patients, adverse drug effects (ADEs) and medication burden associated with TKI therapy can lead to decreased quality of life. Additionally, TKIs have drug interactions that may negatively impact patients' management of co-morbidities or lead to increased ADEs. CASE REPORT: A 65-year-old female with a history of anxiety treated and controlled with venlafaxine experienced increased and resistant anxiety and insomnia after starting dasatinib for CML. MANAGEMENT AND OUTCOME: On dasatinib, the patient experienced worsening anxiety and insomnia. The stress of a new leukemia diagnosis, drug interactions, and ADEs from dasatinib were considered possible causes. Dose adjustments to dasatinib and venlafaxine were made to control the patient's symptoms. However, the patient's symptoms did not resolve. After being on dasatinib for 2.5 years, the patient discontinued TKI therapy due to being in a deep molecular remission and given ongoing challenges managing anxiety. Within 4 months of stopping dasatinib, the patient reported an improvement in anxiety and overall emotional wellbeing. She continues to feel better and remains in a complete molecular remission 20 months off treatment. DISCUSSION: This case demonstrates a possible previously unknown drug interaction with dasatinib as well as a possible rarely reported ADE of dasatinib. Additionally, it highlights the difficulties patients with psychiatric disorders may face on TKI therapy and challenges providers may have in identifying rare psychiatric ADEs, thus emphasizing the need for documentation of these types of cases.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Distúrbios do Início e da Manutenção do Sono , Feminino , Humanos , Idoso , Dasatinibe/efeitos adversos , Cloridrato de Venlafaxina/efeitos adversos , Qualidade de Vida , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Inibidores de Proteínas Quinases/efeitos adversos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico
6.
J Oncol Pharm Pract ; 29(5): 1119-1124, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35491703

RESUMO

INTRODUCTION: In this study, we aim to determine the risk of bleeding or thrombosis with concurrent use of tyrosine kinase inhibitors (TKIs) used to treat CML, and serotonin reuptake inhibitors (SSRIs). METHODS: We conducted a retrospective cohort study of patients with CP-CML cared for at Massachusetts General Hospital (MGH) between April 2016 to February 2021. Participants were included if diagnosed with CP-CML and began TKI treatment (imatinib, dasatinib, nilotinib, bosutinib, or ponatinib) after April 2016. RESULTS: One hundred patients were evaluated, eighty of whom were taking TKIs only (median age 55, 40% female), and twenty were taking TKI and SSRI concomitantly (median age 53.5, 55% female). Baseline demographics between these groups were similar across all variables. Patients in the TKI only group had 9 bleeding events and 3 thrombotic events. Patients in the combination group had 6 bleeding events and 1 thrombotic event. There was no difference between overall rates of major bleeding (4% v. 10%, p = 0.26) or thrombotic events (4% v. 5%, p = 1). However, patients in the combination group were more likely to have major intracranial bleeding events (0% v. 10%, p = 0.04), and there was a trend to significance for minor bleeding events (7.5% v. 20%, p = 0.11). CONCLUSIONS: Concomitant use of TKIs and SSRIs does not appear to increase the total risk of bleeding or thrombotic events compared to patients on TKIs only. However, concomitant use of TKIs and SSRIs may increase risk of intracranial bleeding. Further work is needed to fully assess this risk.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Inibidores Seletivos de Recaptação de Serotonina , Humanos , Feminino , Pessoa de Meia-Idade , Masculino , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Incidência , Estudos Retrospectivos , Dasatinibe/efeitos adversos , Hemorragia/induzido quimicamente , Hemorragia/epidemiologia
7.
Oncologist ; 27(2): 82-86, 2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35641210

RESUMO

BCR-ABL1 kinase inhibitors have improved the prognosis of Philadelphia-chromosome-positive (Ph+)-acute lymphoblastic leukemia (ALL). Ph-like (or BCR-ABL1-like) ALL does not express BCR-ABL1 but commonly harbors other genomic alterations of signaling molecules that may be amenable to therapy. Here, we report a case with a NUP214-ABL1 fusion detected at relapse by multiplexed, targeted RNA sequencing. It had escaped conventional molecular work-up at diagnosis, including cytogenetic analysis and fluorescence in situ hybridization for ABL1 rearrangements. The patient had responded poorly to initial multi-agent chemotherapy and inotuzumab immunotherapy at relapse before the fusion was revealed. The addition of dasatinib targeting NUP214-ABL1 to inotuzumab resulted in complete molecular remission, but recurrence occurred rapidly with dasatinib alone. However, deep molecular remission was recaptured with a combination of blinatumomab and ponatinib, so he could proceed to allotransplantation. This case illustrates that next-generation sequencing approaches designed to discover cryptic gene fusions can benefit patients with Ph-like ALL.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras , Dasatinibe/uso terapêutico , Humanos , Imunoterapia , Hibridização in Situ Fluorescente/métodos , Masculino , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Recidiva
8.
Oncologist ; 27(11): 930-939, 2022 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-35852437

RESUMO

BACKGROUND: Precision oncology relies on molecular diagnostics, and the value-proposition of modern healthcare networks promises a higher standard of care across partner sites. We present the results of a clinical pilot to standardize precision oncology workflows. METHODS: Workflows are defined as the development, roll-out, and updating of disease-specific molecular order sets. We tracked the timeline, composition, and effort of consensus meetings to define the combination of molecular tests. To assess clinical impact, we examined order set adoption over a two-year period (before and after roll-out) across all gastrointestinal and hepatopancreatobiliary (GI) malignancies, and by provider location within the network. RESULTS: Development of 12 disease center-specific order sets took ~9 months, and the average number of tests per indication changed from 2.9 to 2.8 (P = .74). After roll-out, we identified significant increases in requests for GI patients (17%; P < .001), compliance with testing recommendations (9%; P < .001), and the fraction of "abnormal" results (6%; P < .001). Of 1088 GI patients, only 3 received targeted agents based on findings derived from non-recommended orders (1 before and 2 after roll-out); indicating that our practice did not negatively affect patient treatments. Preliminary analysis showed 99% compliance by providers in network sites, confirming the adoption of the order sets across the network. CONCLUSION: Our study details the effort of establishing precision oncology workflows, the adoption pattern, and the absence of harm from the reduction of non-recommended orders. Establishing a modifiable communication tool for molecular testing is an essential component to optimize patient care via precision oncology.


Assuntos
Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/terapia , Medicina de Precisão/métodos , Fluxo de Trabalho , Oncologia/métodos , Atenção à Saúde
9.
J Natl Compr Canc Netw ; 20(9): 1063-1068, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36075385

RESUMO

Interferons are cytokines with immunomodulatory properties that have been used in the treatment of myeloproliferative neoplasms (MPNs) for decades. However, their widespread use has been hampered by their adverse effect profile and difficulty with administration. Recently there has been a resurgence of interest in the use of interferons in MPNs given the development of pegylated formulations with improved tolerability. Currently, treatments for polycythemia vera (PV) and essential thrombocythemia (ET) are targeted toward decreasing the risk of thrombotic complications, because there are no approved therapies that are known to modify disease. However, recent data on interferons in MPNs have suggested the potential for disease-modifying activity, including the achievement of molecular remission and sustained clinical response. This development has led to the question of whether interferons should move forward as the preferred frontline cytoreductive agent for ET and PV, and challenges the criteria currently used to initiate therapy. We review randomized controlled trial data evaluating interferon's efficacy and tolerability in patients with ET and PV. We then consider the data in the context of interferon's known advantages and disadvantages to address whether interferons should be the first choice for cytoreductive treatment in patients with ET and PV.


Assuntos
Transtornos Mieloproliferativos , Policitemia Vera , Trombocitemia Essencial , Procedimentos Cirúrgicos de Citorredução/efeitos adversos , Humanos , Interferons/efeitos adversos , Transtornos Mieloproliferativos/tratamento farmacológico , Policitemia Vera/complicações , Ensaios Clínicos Controlados Aleatórios como Assunto , Trombocitemia Essencial/complicações
10.
J Natl Compr Canc Netw ; 20(9): 1033-1062, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36075392

RESUMO

The classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) consist of myelofibrosis, polycythemia vera, and essential thrombocythemia and are a heterogeneous group of clonal blood disorders characterized by an overproduction of blood cells. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for MPN were developed as a result of meetings convened by a multidisciplinary panel with expertise in MPN, with the goal of providing recommendations for the management of MPN in adults. The Guidelines include recommendations for the diagnostic workup, risk stratification, treatment, and supportive care strategies for the management of myelofibrosis, polycythemia vera, and essential thrombocythemia. Assessment of symptoms at baseline and monitoring of symptom status during the course of treatment is recommended for all patients. This article focuses on the recommendations as outlined in the NCCN Guidelines for the diagnosis of MPN and the risk stratification, management, and supportive care relevant to MF.


Assuntos
Transtornos Mieloproliferativos , Policitemia Vera , Mielofibrose Primária , Trombocitemia Essencial , Adulto , Humanos , Oncologia , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/terapia , Policitemia Vera/diagnóstico , Mielofibrose Primária/diagnóstico , Trombocitemia Essencial/diagnóstico
11.
Cancer ; 127(14): 2500-2506, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33764526

RESUMO

BACKGROUND: Patients with acute myeloid leukemia (AML) receiving intensive chemotherapy face a life-threatening illness, isolating hospitalization, and substantial physical and psychological symptoms. However, data are limited regarding risk factors of posttraumatic stress disorder (PTSD) symptoms in this population. METHODS: The authors conducted a secondary analysis of data from 160 patients with high-risk AML who were enrolled in a supportive care trial. The PTSD Checklist-Civilian Version was used to assess PTSD symptoms at 1 month after AML diagnosis. The Brief COPE and the Functional Assessment of Cancer Therapy-Leukemia were to assess coping and quality of life (QOL), respectively. In addition, multivariate regression models were constructed to assess the relation between PTSD symptoms and baseline sociodemographic factors, coping, and QOL. RESULTS: Twenty-eight percent of patients reported PTSD symptoms, describing high rates of intrusion, avoidance, and hypervigiliance. Baseline sociodemographic factors significantly associated with PTSD symptoms were age (B = -0.26; P = .002), race (B = -8.78; P = .004), and postgraduate education (B = -6.30; P = .029). Higher baseline QOL (B = -0.37; P ≤ .001) and less decline in QOL during hospitalization (B = -0.05; P = .224) were associated with fewer PTSD symptoms. Approach-oriented coping (B = -0.92; P = .001) was associated with fewer PTSD symptoms, whereas avoidant coping (B = 2.42; P ≤ .001) was associated with higher PTSD symptoms. CONCLUSIONS: A substantial proportion of patients with AML report clinically significant PTSD symptoms 1 month after initiating intensive chemotherapy. Patients' baseline QOL, coping strategies, and extent of QOL decline during hospitalization emerge as important risk factors for PTSD, underscoring the need for supportive oncology interventions to reduce the risk of PTSD in this population.


Assuntos
Leucemia Mieloide Aguda , Transtornos de Estresse Pós-Traumáticos , Adaptação Psicológica , Hospitalização , Humanos , Leucemia Mieloide Aguda/complicações , Qualidade de Vida/psicologia , Transtornos de Estresse Pós-Traumáticos/diagnóstico , Transtornos de Estresse Pós-Traumáticos/epidemiologia , Transtornos de Estresse Pós-Traumáticos/etiologia
12.
Oncologist ; 26(11): e2082-e2085, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34272781

RESUMO

Ruxolitinib, a selective inhibitor of Janus kinases 1 and 2, is increasingly being used in allogeneic hematopoietic cell transplantation (HCT) recipients following its approval by the U.S. Food and Drug Administration for the treatment of steroid-refractory acute graft-versus-host disease. Although there is extensive experience using ruxolitinib for patients with myeloproliferative neoplasms, the biologic effects and clinical implications of its dosing, tapering, and discontinuation for allogeneic HCT recipients are incompletely characterized. We describe three allogeneic HCT recipients who developed acute hypoxemic respiratory failure within 3 months of ruxolitinib discontinuation. Radiographic findings included marked bilateral ground-glass opacities. Systemic corticosteroids and reinitiation of ruxolitinib resulted in rapid clinical improvement in all three patients. All three patients achieved a significant clinical response, with decrease in oxygen requirement and improvement in radiographic changes. Given the increasing use of ruxolitinib in allogeneic HCT recipients, there is significant impetus to characterize the biologic and clinical effects resulting from discontinuation of ruxolitinib, to better tailor treatment plans and prevent potential adverse effects.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Insuficiência Respiratória , Humanos , Nitrilas , Pirazóis , Pirimidinas , Estados Unidos , United States Food and Drug Administration
13.
Blood ; 134(25): 2242-2248, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31562135

RESUMO

Recurrent mutations in calreticulin are present in ∼20% of patients with myeloproliferative neoplasms (MPNs). Since its discovery in 2013, we now have a more precise understanding of how mutant CALR, an endoplasmic reticulum chaperone protein, activates the JAK/STAT signaling pathway via a pathogenic binding interaction with the thrombopoietin receptor MPL to induce MPNs. In this Spotlight article, we review the current understanding of the biology underpinning mutant CALR-driven MPNs, discuss clinical implications, and highlight future therapeutic approaches.


Assuntos
Calreticulina , Mutação , Transtornos Mieloproliferativos , Proteínas de Neoplasias , Transdução de Sinais , Animais , Calreticulina/genética , Calreticulina/metabolismo , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Camundongos , Camundongos Transgênicos , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores de Trombopoetina/genética , Receptores de Trombopoetina/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo
14.
Curr Opin Hematol ; 27(2): 122-127, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31904665

RESUMO

PURPOSE OF REVIEW: Atypical chronic myeloid leukemia (aCML), BCR-ABL1-negative, is a rare myelodysplastic/myeloproliferative neoplasm (MDS/MPN) characterized by leukocytosis, granulocytic dysplasia, and typically poor patient outcomes. Since its first description as a variant CML lacking the Philadelphia chromosome (Ph), the diagnostic criteria for aCML have evolved significantly. Nevertheless, distinguishing it from other Ph-negative myeloid neoplasms can still be very challenging, and given its generally worse prognosis, this is a clinically important distinction. The purpose of this review is to conceptualize our understanding of aCML molecular genetics based on recent advances, and describe how genetic features can be used in conjunction with clinical and morphologic features to better diagnose this elusive entity. RECENT FINDINGS: The classification criteria for aCML have evolved and changed multiple times over the past decades, and is now based on strict application of morphologic, clinical and laboratory criteria. Recent work has elucidated the mutational landscape of aCML, especially with respect to potentially differentiating profiles compared with other Ph-negative myeloid neoplasms. SUMMARY: Atypical CML is a rare MDS/MPN overlap syndrome that can be diagnostically challenging; however, its emerging molecular genetic understanding and clinicomorphologic phenotype can help in distinguishing it from other Ph-negative myeloid neoplasms.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/patologia
15.
Cancer ; 126(6): 1264-1273, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31860140

RESUMO

BACKGROUND: Outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) remain poor. Novel therapies specifically targeting AML are of high interest. Brentuximab vedotin (BV) is an antibody-drug conjugate that is specific for human CD30. In this phase 1 dose escalation study, the authors evaluated the safety of BV combined with mitoxantrone, etoposide, and cytarabine (MEC) re-induction chemotherapy for patients with CD30-expressing R/R AML. METHODS: Using a standard dose escalation design, the authors evaluated 3 dose levels of BV (0.9 mg/kg, 1.2 mg/kg, and 1.8 mg/kg) administered once on day 1 followed by MEC on days 3 through 7. RESULTS: There were no dose-limiting toxicities noted and the maximum tolerated dose was not reached. The recommended phase 2 dose of BV was determined to be 1.8 mg/kg when combined with MEC. The side effect profile was similar to that expected from MEC chemotherapy alone, with the most common grade ≥3 toxicities being febrile neutropenia, thrombocytopenia, and anemia (toxicities were graded using the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). Among the 22 patients enrolled on the trial, the composite response rate was 36%, with a composite response rate of 42% noted among those who received the highest dose of BV. The median overall survival was 9.5 months, with a median disease-free survival of 6.8 months observed among responders. Approximately 55% of patients were able to proceed with either allogeneic hematopoietic stem cell transplantation or donor lymphocyte infusion. CONCLUSIONS: The combination of BV with MEC was found to be safe in patients with CD30-expressing R/R AML and warrants further study comparing this combination with the use of MEC alone in this population (ClinicalTrials.gov identifier NCT01830777). LAY SUMMARY: The outcomes for patients with relapsed/refractory acute myeloid leukemia (R/R AML) are exceptionally poor. New and emerging treatment combinations are actively being studied in an effort to improve outcomes. The authors examined the combination of brentuximab vedotin, an antibody product that recognizes a marker called CD30, with mitoxantrone, etoposide, and cytarabine (MEC), a common chemotherapy regimen, in patients with R/R AML that expressed the CD30 marker. The authors found that the combination was safe and well tolerated. Future studies comparing this new combination with the use of MEC alone can help to inform its effectiveness for this patient population.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Brentuximab Vedotin/administração & dosagem , Imunoconjugados/administração & dosagem , Quimioterapia de Indução/métodos , Leucemia Mieloide Aguda/tratamento farmacológico , Adulto , Idoso , Antineoplásicos Imunológicos/efeitos adversos , Brentuximab Vedotin/efeitos adversos , Citarabina/administração & dosagem , Intervalo Livre de Doença , Esquema de Medicação , Resistencia a Medicamentos Antineoplásicos , Etoposídeo/administração & dosagem , Feminino , Humanos , Imunoconjugados/efeitos adversos , Antígeno Ki-1/metabolismo , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/mortalidade , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Mitoxantrona/administração & dosagem , Recidiva , Adulto Jovem
16.
J Natl Compr Canc Netw ; 18(9): 1271-1278, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32886896

RESUMO

Primary myelofibrosis (PMF) has the least favorable prognosis of the Philadelphia chromosome-negative myeloproliferative neoplasms, which also include essential thrombocythemia (ET) and polycythemia vera (PV). However, clinical presentations and outcomes of PMF vary widely, with median overall survival ranging from years to decades. Given the heterogeneity of PMF, there has been considerable effort to develop discriminatory prognostic models to help with management decisions, particularly for the consideration of hematopoietic stem cell transplantation in patients at higher risk. Although earlier models incorporated only clinical features in risk stratification, contemporary models increasingly use molecular and cytogenetic features, leading to more comprehensive prognostication. This article reviews the most widely adopted prognostic models used for PMF, including the International Prognostic Scoring System (IPSS), dynamic IPSS (DIPSS)/DIPSS+, mutation-enhanced IPSS for transplant-age patients (MIPSS70)/MIPSS70+/MIPSS70+ version 2.0, genetically inspired prognostic scoring system, and Myelofibrosis Secondary to PV and ET-Prognostic Model in patients with post-ET/PV myelofibrosis. We also discuss newly emerging prognostic models and provide a practical approach to risk stratification in patients with PMF and post-ET/PV myelofibrosis.


Assuntos
Transtornos Mieloproliferativos , Policitemia Vera , Mielofibrose Primária , Trombocitemia Essencial , Humanos , Mielofibrose Primária/diagnóstico , Mielofibrose Primária/genética , Mielofibrose Primária/terapia , Prognóstico
17.
J Natl Compr Canc Netw ; 18(10): 1385-1415, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33022644

RESUMO

Chronic myeloid leukemia (CML) is defined by the presence of Philadelphia chromosome (Ph) which results from a reciprocal translocation between chromosomes 9 and 22 [t(9;22] that gives rise to a BCR-ABL1 fusion gene. CML occurs in 3 different phases (chronic, accelerated, and blast phase) and is usually diagnosed in the chronic phase. Tyrosine kinase inhibitor therapy is a highly effective first-line treatment option for all patients with newly diagnosed chronic phase CML. This manuscript discusses the recommendations outlined in the NCCN Guidelines for the diagnosis and management of patients with chronic phase CML.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , Leucemia Mieloide de Fase Crônica , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Oncologia , Cromossomo Filadélfia , Translocação Genética
18.
J Natl Compr Canc Netw ; 18(9): 1248-1269, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32886902

RESUMO

Eosinophilic disorders and related syndromes represent a heterogeneous group of neoplastic and nonneoplastic conditions, characterized by more eosinophils in the peripheral blood, and may involve eosinophil-induced organ damage. In the WHO classification of myeloid and lymphoid neoplasms, eosinophilic disorders characterized by dysregulated tyrosine kinase (TK) fusion genes are recognized as a new category termed, myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1 or with PCM1-JAK2. In addition to these aforementioned TK fusion genes, rearrangements involving FLT3 and ABL1 genes have also been described. These new NCCN Guidelines include recommendations for the diagnosis, staging, and treatment of any one of the myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) and a TK fusion gene included in the 2017 WHO Classification, as well as MLN-Eo and a FLT3 or ABL1 rearrangement.


Assuntos
Eosinofilia , Transtornos Mieloproliferativos , Neoplasias , Eosinofilia/diagnóstico , Eosinofilia/genética , Humanos , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/terapia , Proteínas de Fusão Oncogênica/genética
19.
Cancer ; 125(21): 3845-3852, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31299106

RESUMO

BACKGROUND: Older adults with acute myeloid leukemia (AML) are often assumed to have poor outcomes after admission to the intensive care unit (ICU). However, little is known about ICU utilization and post-ICU outcomes in this population. METHODS: The authors conducted a retrospective analysis for 330 patients who were 60 years old or older and were diagnosed with AML between 2005 and 2013 at 2 hospitals in Boston.They used descriptive statistics to examine the proportion of patients admitted to the ICU as well as their mortality and functional recovery. They used logistic regression to identify risk factors for in-hospital mortality. RESULTS: Ninety-six patients (29%) were admitted to the ICU, primarily because of respiratory failure (39%), septic shock (28%), and neurological compromise (9%). The proportions of patients who survived to hospital discharge, 90 days, and 1 year were 47% (45 of 96), 35% (34 of 96), and 30% (29 of 96), respectively. At 90 days, 76% of the patients had an Eastern Cooperative Oncology Group performance status (ECOG PS) of 0 or 1, and 86% were in complete remission (CR) and/or continued to receive AML-directed therapy. In a multivariate analysis, a poorer baseline ECOG PS score (odds ratio, 2.76; P = .013) and the need for 2 or more life-sustaining therapies (ie, vasopressors, invasive ventilation, and/or renal replacement therapy; odds ratio, 12.4; P < .001) were associated with increased odds of in-hospital mortality. CONCLUSIONS: Although almost one-third of older patients with AML are admitted to an ICU, nearly half survive to hospital discharge with good functional outcomes. The baseline performance status and the need for 2 or more life-sustaining therapies predict hospital mortality. These data support the judicious use of ICU resources for older patients with AML.


Assuntos
Mortalidade Hospitalar/tendências , Hospitalização/estatística & dados numéricos , Unidades de Terapia Intensiva/estatística & dados numéricos , Leucemia Mieloide/terapia , Doença Aguda , Idoso , Boston , Feminino , Humanos , Leucemia Mieloide/diagnóstico , Leucemia Mieloide/mortalidade , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde , Prognóstico , Estudos Retrospectivos
20.
Cancer ; 125(4): 541-549, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30422308

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) cells harboring mutations in isocitrate dehydrogenase 1 (IDH1) and isocitrate dehydrogenase 2 (IDH2) produce the oncometabolite 2-hydroxyglutarate (2HG). This study prospectively evaluated the 2HG levels, IDH1/2 mutational status, and outcomes of patients receiving standard chemotherapy for newly diagnosed AML. METHODS: Serial samples of serum, urine, and bone marrow aspirates were collected from patients newly diagnosed with AML, and 2HG levels were measured with mass spectrometry. Patients with baseline serum 2HG levels greater than 1000 ng/mL or marrow pellet 2HG levels greater than 1000 ng/2 × 106 cells, which suggested the presence of an IDH1/2 mutation, underwent serial testing. IDH1/2 mutations and estimated variant allele frequencies were identified. AML characteristics were compared with the Wilcoxon test and Fisher's exact test. Disease-free survival and overall survival (OS) were evaluated with log-rank tests and Cox regression. RESULTS: Two hundred and two patients were treated for AML; 51 harbored IDH1/2 mutations. IDH1/2-mutated patients had significantly higher 2HG levels in serum, urine, bone marrow aspirates, and aspirate cell pellets than wild-type patients. A serum 2HG level greater than 534.5 ng/mL was 98.8% specific for the presence of an IDH1/2 mutation. Patients with IDH1/2-mutated AML treated with 7+3-based induction had a 2-year event-free survival (EFS) rate of 44% and a 2-year OS rate of 57%. There was no difference in complete remission rates, EFS, or OS between IDH1/2-mutated and wild-type patients. Decreased serum 2HG levels on day 14 as a proportion of the baseline were significantly associated with improvements in EFS (P = .047) and OS (P = .019) in a multivariate analysis. CONCLUSIONS: Among patients with IDH1/2-mutated AML, 2HG levels are highly specific for the mutational status at diagnosis, and they have prognostic relevance in patients receiving standard chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/genética , Glutaratos/sangue , Isocitrato Desidrogenase/genética , Leucemia Mieloide Aguda/mortalidade , Mutação , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Taxa de Sobrevida , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA