Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Blood Adv ; 8(6): 1515-1528, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38290135

RESUMO

ABSTRACT: Ruxolitinib reduces spleen volume, improves symptoms, and increases survival in patients with intermediate- or high-risk myelofibrosis. However, suboptimal response may occur, potentially because of signaling via the phosphoinositide 3-kinase (PI3K)/protein kinase B pathway. This phase 2 study evaluated dosing, efficacy, and safety of add-on PI3Kδ inhibitor parsaclisib for patients with primary or secondary myelofibrosis with suboptimal response to ruxolitinib. Eligible patients remained on a stable ruxolitinib dose and received add-on parsaclisib 10 or 20 mg, once daily for 8 weeks, and once weekly thereafter (daily-to-weekly dosing; n = 32); or parsaclisib 5 or 20 mg, once daily for 8 weeks, then 5 mg once daily thereafter (all-daily dosing; n = 42). Proportion of patients achieving a ≥10% decrease in spleen volume at 12 weeks was 28% for daily-to-weekly dosing and 59.5% for all-daily dosing. Proportions of patients achieving ≥50% decrease at week 12 in Myelofibrosis Symptom Assessment Form and Myeloproliferative Neoplasms Symptom Assessment Form symptom scores were 14% and 18% for daily-to-weekly dosing, and 28% and 32% for all-daily dosing, respectively. Most common nonhematologic treatment-emergent adverse events were nausea (23%), diarrhea (22%), abdominal pain and fatigue (each 19%), and cough and dyspnea (each 18%). New-onset grade 3 and 4 thrombocytopenia were observed in 19% of patients, each dosed daily-to-weekly, and in 26% and 7% of patients dosed all-daily, respectively, managed with dose interruptions. Hemoglobin levels remained steady. The addition of parsaclisib to stable-dose ruxolitinib can reduce splenomegaly and improve symptoms, with manageable toxicity in patients with myelofibrosis with suboptimal response to ruxolitinib. This trial was registered at www.clinicaltrials.gov as #NCT02718300.


Assuntos
Nitrilas , Mielofibrose Primária , Pirimidinas , Pirrolidinas , Humanos , Mielofibrose Primária/diagnóstico , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/induzido quimicamente , Fosfatidilinositol 3-Quinases , Pirazóis/efeitos adversos
2.
HERD ; 17(2): 183-199, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38166516

RESUMO

OBJECTIVE: To understand parent and child perception of spaces experienced during outpatient procedures and to measure their anxiety in these spaces. BACKGROUND: Same-day procedures are becoming prevalent among children in the United States. While studies conducted in different types of healthcare settings show that the physical environment influences healthcare experiences of patients, there is a lack of research on patient and family perceptions of the physical environment of the outpatient centers where such procedures are conducted. METHODS: This study used ecological momentary assessment to collect patient experience and anxiety data at different points during the patient's journey through an ambulatory surgical center where pediatric gastrointestinal (GI) procedures were performed. Objective and subjective measures of anxiety were collected. A Qualtrics survey asked participants' perceptions about four spaces-waiting, preprocedure, procedure, and recovery. RESULTS: Child participants reported liking murals, double chairs, patient beds, wall color, and access to a television. They disliked medical equipment and lack of child-friendly furniture. Most parents liked the murals, access to a television, and nature photos, while disliking the lack of privacy, lack of toys in waiting areas, and lack of child-friendly furniture. On average, both children and parents experienced the highest anxiety levels before and during the procedure and the lowest during recovery. Between the four spaces, no significant differences were observed in the heart rate variability and skin conductance responses for both groups. CONCLUSIONS: Despite the outpatient nature of the procedures, participants experienced anxiety before the GI procedure. Comfortable design features that provide distractions are preferred by children and their parents.


Assuntos
Ansiedade , Decoração de Interiores e Mobiliário , Pais , Humanos , Projetos Piloto , Pais/psicologia , Criança , Masculino , Feminino , Pré-Escolar , Procedimentos Cirúrgicos Ambulatórios/psicologia , Avaliação Momentânea Ecológica , Adolescente , Inquéritos e Questionários , Adulto , Instituições de Assistência Ambulatorial , Pacientes Ambulatoriais/psicologia , Percepção
3.
Leuk Lymphoma ; 64(10): 1662-1672, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37424335

RESUMO

Even though overexpression of the immune checkpoint protein, programmed cell death ligand-1 (PD-L1), is observed in solid tumors, its expression patterns in acute myeloid leukemia remain understudied. As activation of the JAK/STAT pathway has been shown to enhance PD-L1 expression in preclinical models, we evaluated biopsies from AML patients with activating mutations in JAK2/STATs. PD-L1 expression was significantly upregulated in JAK2/STAT mutant cases when compared to JAK2 wildtype controls as demonstrated by PD-L1 immunohistochemistry staining and quantified using the combined positive score (CPS) system. There is significant overexpression of phosphorylated STAT3 expression in patients with oncogenic JAK2 activation and a positive correlation between p-STAT3 and PD-L1 expression. In conclusion, we demonstrate the CPS scoring system could be applied as a quantitative measure of PD-L1 expression in leukemias and that JAK2/STATs mutant AML can be potential candidates for checkpoint inhibitor trials.


Assuntos
Antígeno B7-H1 , Leucemia Mieloide Aguda , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Janus Quinases/genética , Janus Quinases/metabolismo , Leucemia Mieloide Aguda/genética , Mutação , Transdução de Sinais/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Regulação para Cima
5.
Clin Cancer Res ; 29(5): 943-956, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36537918

RESUMO

PURPOSE: The Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) polycythemia vera, essential thrombocythemia, and primary myelofibrosis are characterized by JAK/STAT pathway activation. JAK inhibitors are approved for MPN treatment, but persistence has been observed, due to JAK/STAT reactivation. EXPERIMENTAL DESIGN: Using MPN patient samples, JAK2-mutated cell lines, and MPN mouse models, we examined both the efficacy and mechanism by which crizotinib, the ALK/MET/RON/ROS1 inhibitor approved for the treatment of non-small cell lung cancer, alters MPN cell proliferation and JAK/STAT activation. RESULTS: We found that crizotinib suppresses proliferation and activation of JAK/STAT signaling, and decreases the disease burden in the JAK2V617F mouse model of MPN. Furthermore, we found that crizotinib could overcome JAK inhibitor persistence to ruxolitinib. Interestingly, phosphorylation of the crizotinib target RON kinase was enhanced in ruxolitinib-persistent cells. We show that phospho-JAK2 and phospho-RON can physically interact to sustain JAK/STAT signaling, and that the combination of crizotinib and ruxolitinib disrupts this interaction. Furthermore, RON knockdown suppresses proliferation and activation of JAK/STAT signaling in JAK2-mutated cells, and RON deletion in a JAK2V617F mouse MPN model decreases the disease burden. We also observed RON hyperactivation in MPN patient cells, suggesting that RON may be an important target of crizotinib in MPN. CONCLUSIONS: In summary, we demonstrate that crizotinib has preclinical efficacy in MPN patient cells, JAK2-mutated cell lines, and a JAK2-mutated mouse model, and that the combination of crizotinib with JAK inhibitors suppresses JAK inhibitor persistence. Our work suggests that crizotinib should be investigated for the treatment of patients with MPN.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Inibidores de Janus Quinases , Neoplasias Pulmonares , Transtornos Mieloproliferativos , Animais , Camundongos , Inibidores de Janus Quinases/uso terapêutico , Crizotinibe/farmacologia , Crizotinibe/uso terapêutico , Janus Quinases/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Transdução de Sinais , Fatores de Transcrição STAT/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Janus Quinase 2/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Mutação
6.
Cancer Cell ; 40(1): 3-5, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34838186
7.
Res Pract Thromb Haemost ; 5(6): e12574, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34532628

RESUMO

BACKGROUND: Patients with myeloproliferative neoplasms (MPNs), such as polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are at an increased risk of recurrent thromboembolic events (TEs) and hemorrhagic complications. Anticoagulation with vitamin K antagonists (VKAs) had been the standard of care until the recent US Food and Drug Administration approval of direct oral anticoagulants (DOACs) for treatment of cancer-associated thrombosis. However, since patients with MPNs were underrepresented in large studies, the use of DOACs in patients with MPN-associated thrombosis remains understudied. OBJECTIVES: The primary objective of this study was to establish the incidence of recurrent TEs and hemorrhagic complications in patients with MPN-associated thrombosis treated with DOACs versus VKAs as first-line therapy. METHODS: Data from 30 patients ≥18 years old with established diagnoses of PV or ET who were treated with either DOACs or VKAs as the first-line anticoagulant for arterial and/or venous thrombosis were reviewed to determine the incidence of recurrent TEs as well as hemorrhagic complications. RESULTS: Nineteen patients were treated with DOACs, and 11 were treated with VKAs. Of those on DOACs, 1 had a recurrent thrombosis, and 4 had bleeding events. Of the 11 patients treated with VKAs, 1 had a recurrent thrombotic event, and 1 had a bleeding event. CONCLUSION: Our data did not demonstrate a significant difference in recurrent TEs or bleeding events in patients with MPN-associated thrombosis anticoagulated with either DOACs or VKAs.

9.
Leuk Lymphoma ; 62(1): 167-175, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32924682

RESUMO

p53 together with its downstream product p21 plays an important role in tumorigenesis development. MDM2 and MDM4 are two p53 regulators. We studied the expression of p53, p21, MDM2, and MDM4 in a total of 120 cases of myeloid neoplasms including MDS, AML or MDS/MPN, and control, using single and double immunohistochemical stains. We found TP53 mutations had a worse outcome in patients with AML/MDS, and p53 expression detected by immunohistochemistry had a similar prognostic value. p21 expression was strongly related to TP53 mutation status, with loss of expression in almost all TP53 mutated cases. MDM2 and MDM4 were highly expressed in hematopoietic cells in both benign and neoplastic cells. MDM2/p53 double positive cells exceeded MDM4/p53 double positive cells in neoplastic cases. Finally, we observed that p21 protein expression was up regulated upon the use of ALRN-6924 (Aileron) while no significant changes were seen in p53, MDM2 and MDM4 expression.


Assuntos
Neoplasias , Proteína Supressora de Tumor p53 , Proteínas de Ciclo Celular/genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética
10.
Blood Adv ; 4(18): 4282-4291, 2020 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-32915978

RESUMO

Momelotinib (MMB) is a JAK1/2 and ACVR1 inhibitor with demonstrated clinical activity in all 3 hallmarks of myelofibrosis (MF): anemia, constitutional symptoms, and splenomegaly. In this phase 2 open-label translational biology study (NCT02515630) of 41 transfusion-dependent patients with MF, we explored mechanisms underlying the favorable activity of MMB on MF-associated iron-restricted anemia, including its impact on serum hepcidin levels, and markers of iron storage and availability, erythropoiesis, and inflammation. A transfusion-independent response (TI-R), defined as red blood cell transfusion independence (TI) ≥12 weeks at any time on study, occurred in 17 patients (41%; 95% confidence interval [CI], 26%-58%), including 14 patients (34%; 95% CI, 20%-51%) who achieved TI-R by week 24. In addition, 78% of TI nonresponse (TI-NR) patients achieved a ≥50% decrease in transfusion requirement for ≥8 weeks. Adverse events (AEs) were consistent with previous studies of MMB in MF, with cough, diarrhea, and nausea as the most common. Twenty-one patients experienced grade ≥3 AEs, most commonly anemia and neutropenia. Consistent with preclinical data, daily MMB treatment led to an acute and persistent decrease in blood hepcidin associated with increased iron availability and markers of erythropoiesis. Baseline characteristics associated with TI-R were lower inflammation and hepcidin as well as increased markers of erythropoiesis and bone marrow function. Overall, the study demonstrates that MMB treatment decreases hepcidin in conjunction with improving iron metabolism and erythropoiesis, suggesting a mechanistic explanation for the reduced transfusion dependency observed in transfusion-dependent MF patients treated with MMB, thereby addressing the key unmet medical need in the MF population.


Assuntos
Mielofibrose Primária , Receptores de Ativinas Tipo I , Benzamidas , Hepcidinas , Humanos , Janus Quinase 1 , Janus Quinase 2/genética , Mielofibrose Primária/tratamento farmacológico , Pirimidinas
12.
Clin Lymphoma Myeloma Leuk ; 20(10): 697-703.e1, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32624445

RESUMO

INTRODUCTION: Ruxolitinib is approved for the treatment of polycythemia vera (PV) with hydroxyurea resistance or intolerance. Approval was based on the phase III RESPONSE trial, which demonstrated efficacy in a highly selected patient population. MATERIALS AND METHODS: To characterize the tolerability and outcomes of ruxolitinib outside of a clinical trial, we performed a multi-center retrospective analysis of patients with PV treated with ruxolitinib at 11 participating sites across the United States. Outcomes of interest included change in phlebotomy requirements after starting ruxolitinib and spleen response, as these were included in the primary composite outcome in the RESPONSE trial. RESULTS: One hundred twenty-six patients met eligibility criteria, and the median duration of follow-up was 22.4 months (range, 0-63.0 months). At 32 weeks after starting ruxolitinib, the percentage of patients who received at least 1 phlebotomy was significantly decreased compared with before ruxolitinib (37% vs. 56%; relative risk [RR], 0.66; 95% confidence interval [CI], 0.52-0.84; P < .001). Phlebotomy requirements were similarly decreased in patients who had received at least 3 phlebotomies prior to ruxolitinib initiation (28% vs. 17%; RR, 1.65; 95% CI, 1.13-2.40; P < .01). Resolution of palpable splenomegaly was also documented (48% vs. 20%; RR, 2.45; 95% CI, 1.70-3.53; P < .0001). A total of 9.5% of patients discontinued ruxolitinib owing to treatment-emergent adverse events, and 81.7% of patients were receiving ruxolitinib at last known follow-up. CONCLUSION: These real-world results are similar to those reported from the RESPONSE trial, although additional follow-up is necessary to assess long-term outcomes and potential for late-onset toxicity.


Assuntos
Janus Quinases/uso terapêutico , Policitemia Vera/tratamento farmacológico , Pirazóis/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Janus Quinases/farmacologia , Masculino , Pessoa de Meia-Idade , Nitrilas , Pirazóis/farmacologia , Pirimidinas , Resultado do Tratamento , Adulto Jovem
14.
Blood ; 132(14): 1507-1518, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30104217

RESUMO

Adult T-cell leukemia lymphoma (ATLL) is a rare T cell neoplasm that is endemic in Japanese, Caribbean, and Latin American populations. Most North American ATLL patients are of Caribbean descent and are characterized by high rates of chemo-refractory disease and worse prognosis compared with Japanese ATLL. To determine genomic differences between these 2 cohorts, we performed targeted exon sequencing on 30 North American ATLL patients and compared the results with the Japanese ATLL cases. Although the frequency of TP53 mutations was comparable, the mutation frequency in epigenetic and histone modifying genes (57%) was significantly higher, whereas the mutation frequency in JAK/STAT and T-cell receptor/NF-κB pathway genes was significantly lower. The most common type of epigenetic mutation is that affecting EP300 (20%). As a category, epigenetic mutations were associated with adverse prognosis. Dissimilarities with the Japanese cases were also revealed by RNA sequencing analysis of 9 primary patient samples. ATLL samples with a mutated EP300 gene have decreased total and acetyl p53 protein and a transcriptional signature reminiscent of p53-mutated cancers. Most importantly, decitabine has highly selective single-agent activity in the EP300-mutated ATLL samples, suggesting that decitabine treatment induces a synthetic lethal phenotype in EP300-mutated ATLL cells. In conclusion, we demonstrate that North American ATLL has a distinct genomic landscape that is characterized by frequent epigenetic mutations that are targetable preclinically with DNA methyltransferase inhibitors.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Decitabina/uso terapêutico , Leucemia-Linfoma de Células T do Adulto/tratamento farmacológico , Leucemia-Linfoma de Células T do Adulto/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/efeitos dos fármacos , Proteína p300 Associada a E1A/genética , Epigênese Genética , Feminino , Humanos , Japão/epidemiologia , Leucemia-Linfoma de Células T do Adulto/diagnóstico , Leucemia-Linfoma de Células T do Adulto/epidemiologia , Masculino , Pessoa de Meia-Idade , Taxa de Mutação , Prognóstico , Transcriptoma , Proteína Supressora de Tumor p53/genética , Estados Unidos/epidemiologia
15.
Anticancer Res ; 38(4): 2201-2205, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29599340

RESUMO

BACKGROUND: Sweet's syndrome (SS) is a febrile neutrophilic dermatosis that has been clinically linked to hematological malignancies, particularly myelodysplastic syndrome (MDS), in a number of case series. Many epigenetic changes underlying MDS have been identified, such as a mutation in the isocitrate dehydrogenase 1 (IDH1) gene, which causes DNA hypermethylation and alteration of a number of genes that lead to leukemogenesis. However, the pathogenesis of malignancy-associated SS is unknown. CASE REPORT: We present two patients who were diagnosed with SS and concomitant IDH1-mutated MDS. Immunohistochemical staining of their skin lesions showed neutrophils diffusely positive for the IDH1 mutation. CONCLUSION: These cases demonstrate that IDH1 mutation may be implicated in the pathogenesis of malignancy-associated SS. Future investigation to elucidate this pathway is warranted. Establishing this molecular link can provide an earlier identification of patients with SS who are also at increased risk for developing MDS.


Assuntos
Isocitrato Desidrogenase/genética , Mutação de Sentido Incorreto , Síndromes Mielodisplásicas/genética , Síndrome de Sweet/genética , Idoso , Metilação de DNA , Análise Mutacional de DNA , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/epidemiologia , Polimorfismo de Nucleotídeo Único , Síndrome de Sweet/epidemiologia
16.
J Invest Dermatol ; 138(5): 1116-1125, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29128259

RESUMO

Cutaneous T-cell lymphoma is a heterogeneous group of lymphomas characterized by the accumulation of malignant T cells in the skin. The molecular and cellular etiology of this malignancy remains enigmatic, and what role antigenic stimulation plays in the initiation and/or progression of the disease remains to be elucidated. Deep sequencing of the tumor genome showed a highly heterogeneous landscape of genetic perturbations, and transcriptome analysis of transformed T cells further highlighted the heterogeneity of this disease. Nonetheless, using data harvested from high-throughput transcriptional profiling allowed us to develop a reliable signature of this malignancy. Focusing on a key cytokine signaling pathway previously implicated in cutaneous T-cell lymphoma pathogenesis, JAK/STAT signaling, we used conditional gene targeting to develop a fully penetrant small animal model of this disease that recapitulates many key features of mycosis fungoides, a common variant of cutaneous T-cell lymphoma. Using this mouse model, we show that T-cell receptor engagement is critical for malignant transformation of the T lymphocytes and that progression of the disease is dependent on microbiota.


Assuntos
Citocinas/fisiologia , Linfoma Cutâneo de Células T/etiologia , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/etiologia , Animais , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Linfoma Cutâneo de Células T/genética , Linfoma Cutâneo de Células T/imunologia , Camundongos , Microbiota , Receptores de Antígenos de Linfócitos T/fisiologia , Fator de Transcrição STAT3/fisiologia , Síndrome de Sézary/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia
17.
J Clin Diagn Res ; 11(6): ZJ01-ZJ02, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28764314
18.
Eur J Haematol ; 99(1): 36-41, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28370510

RESUMO

OBJECTIVE: To determine the financial and psycho-social impact of participation in clinical trials of patients with BCR/ABL-negative myeloproliferative neoplasms (MPN). METHODS: An international, observational cross-sectional study was performed in 143 consecutive MPN patients (51% myelofibrosis, 36% polycythemia vera, 13% essential thrombocythemia), 68% from Italy, 17% from USA, and 15% from Spain. RESULTS: Thirty-five percent of patients reported having spent more money during the trial than in previous treatments and 21% having missed more workdays. Twelve percent replied that they would not have participated in the trial if the financial consequences would have been known beforehand. In 10% of the patients, the interpersonal relationships were more affected during the trial than in previous treatment but, overall, 91% subjects believed that participating in the clinical trial was worth the financial or emotional suffering. Concerning patients' suggestions, 54% of them indicated that the number of visits required for the trial should be clearly specified in the informed consent, 60% recommended travel cost reimbursement, and 23% hotel cost reimbursement. CONCLUSIONS: Physicians and pharmaceutical companies involved in clinical trials with patients with hematological diseases should be aware of these problems and make efforts to attenuate the socioeconomic burden of participation in the trials.


Assuntos
Efeitos Psicossociais da Doença , Transtornos Mieloproliferativos/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Ensaios Clínicos como Assunto , Estudos Transversais , Feminino , Humanos , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/terapia , Fatores Socioeconômicos , Espanha/epidemiologia , Estados Unidos/epidemiologia , Adulto Jovem
19.
Blood Adv ; 1(24): 2120-2123, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29296859

RESUMO

SEER data and a Bronx validation cohort demonstrate that Hispanics present with AML at younger age but have shorter survival than whites.Increased frequency of high-risk mutations in Hispanics provides a potential biologic explanation for poorer outcomes in Hispanics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA