Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Blood ; 143(21): 2190-2200, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38306657

RESUMO

ABSTRACT: VEXAS (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) syndrome, caused by somatic mutations in UBA1, is an autoinflammatory disorder with diverse systemic manifestations. Thrombosis is a prominent clinical feature of VEXAS syndrome. The risk factors and frequency of thrombosis in VEXAS syndrome are not well described, due to the disease's recent discovery and the paucity of large databases. We evaluated 119 patients with VEXAS syndrome for venous and arterial thrombosis and correlated their presence with clinical outcomes and survival. Thrombosis occurred in 49% of patients, mostly venous thromboembolism (VTE; 41%). Almost two-thirds of VTEs were unprovoked, 41% were recurrent, and 20% occurred despite anticoagulation. The cumulative incidence of VTE was 17% at 1 year from symptom onset and 40% by 5 years. Cardiac and pulmonary inflammatory manifestations were associated with time to VTE. M41L was positively associated specifically with pulmonary embolism by univariate (odds ratio [OR]: 4.58, confidence interval [CI] 1.28-16.21, P = .02) and multivariate (OR: 16.94, CI 1.99-144.3, P = .01) logistic regression. The cumulative incidence of arterial thrombosis was 6% at 1 year and 11% at 5 years. The overall survival of the entire patient cohort at median follow-up time of 4.8 years was 88%, and there was no difference in survival between patients with or without thrombosis (P = .8). Patients with VEXAS syndrome are at high risk of VTE; thromboprophylaxis should administered be in high-risk settings unless strongly contraindicated.


Assuntos
Trombose , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Trombose/etiologia , Trombose/genética , Trombose/epidemiologia , Adolescente , Enzimas Ativadoras de Ubiquitina/genética , Adulto Jovem , Fatores de Risco , Idoso , Criança , Trombose Venosa/etiologia , Trombose Venosa/epidemiologia , Trombose Venosa/genética , Incidência , Mutação , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/complicações , Pré-Escolar
2.
Blood ; 142(25): 2146-2158, 2023 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-37738626

RESUMO

ABSTRACT: Deleterious germ line RUNX1 variants cause the autosomal dominant familial platelet disorder with associated myeloid malignancy (FPDMM), characterized by thrombocytopenia, platelet dysfunction, and a predisposition to hematologic malignancies (HMs). We launched a FPDMM natural history study and, from January 2019 to December 2021, enrolled 214 participants, including 111 patients with 39 different RUNX1 variants from 45 unrelated families. Seventy of 77 patients had thrombocytopenia, 18 of 18 had abnormal platelet aggregometry, 16 of 35 had decreased platelet dense granules, and 28 of 55 had abnormal bleeding scores. Nonmalignant bone marrows showed increased numbers of megakaryocytes in 12 of 55 patients, dysmegakaryopoiesis in 42 of 55, and reduced cellularity for age in 30 of 55 adult and 17 of 21 pediatric cases. Of 111 patients, 19 were diagnosed with HMs, including myelodysplastic syndrome, acute myeloid leukemia, chronic myelomonocytic leukemia, acute lymphoblastic leukemia, and smoldering myeloma. Of those 19, 18 were relapsed or refractory to upfront therapy and referred for stem cell transplantation. In addition, 28 of 45 families had at least 1 member with HM. Moreover, 42 of 45 patients had allergic symptoms, and 24 of 30 had gastrointestinal (GI) symptoms. Our results highlight the importance of a multidisciplinary approach, early malignancy detection, and wider awareness of inherited disorders. This actively accruing, longitudinal study will genotype and phenotype more patients with FPDMM, which may lead to a better understanding of the disease pathogenesis and clinical course, which may then inform preventive and therapeutic interventions. This trial was registered at www.clinicaltrials.gov as #NCT03854318.


Assuntos
Neoplasias Hematológicas , Leucemia Mieloide Aguda , Transtornos Mieloproliferativos , Trombocitopenia , Adulto , Humanos , Criança , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Estudos Longitudinais , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/complicações , Trombocitopenia/genética , Transtornos Mieloproliferativos/complicações , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/complicações
3.
Oncologist ; 26(4): 288-e541, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33554406

RESUMO

LESSONS LEARNED: Despite the initial optimism for using immune checkpoint inhibition in the treatment of multiple myeloma, subsequent clinical studies have been disappointing. Preclinical studies have suggested that priming the immune system with various modalities in addition to checkpoint inhibition may overcome the relative T-cell exhaustion or senescence; however, in this small data set, radiotherapy with checkpoint inhibition did not appear to activate the antitumor immune response. BACKGROUND: Extramedullary disease (EMD) is recognized as an aggressive subentity of multiple myeloma (MM) with a need for novel therapeutic approaches. We therefore designed a proof-of-principle pilot study to evaluate the synergy between the combination of the anti-PD-L1, avelumab, and concomitant hypofractionated radiotherapy. METHODS: This was a single-arm phase II Simon two-stage single center study that was prematurely terminated because of the COVID-19 pandemic after enrolling four patients. Key eligibility included patients with relapsed/refractory multiple myeloma (RRMM) who had exhausted or were not candidates for standard therapy and had at least one lesion amenable to radiotherapy. Patients received avelumab until progression or intolerable toxicity and hypofractionated radiotherapy to a focal lesion in cycle 2. Radiotherapy was delayed until cycle 2 to allow the avelumab to reach a study state, given the important observation from previous studies that concomitant therapy is needed for the abscopal effect. RESULTS: At a median potential follow-up of 10.5 months, there were no objective responses, one minimal response, and two stable disease as best response. The median progression-free survival (PFS) was 5.3 months (95% confidence interval [CI]: 2.5-7.1 months), and no deaths occurred. There were no grade ≥3 and five grade 1-2 treatment-related adverse events. CONCLUSION: Avelumab in combination with radiotherapy for patients with RRMM and EMD was associated with very modest systemic clinical benefit; however, patients did benefit as usual from local radiotherapy. Furthermore, the combination was very well tolerated compared with historical RRMM treatment regimens.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Mieloma Múltiplo , Idoso , Idoso de 80 Anos ou mais , COVID-19 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/radioterapia , Pandemias , Projetos Piloto
4.
Transfusion ; 61(4): 1041-1046, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33528026

RESUMO

BACKGROUND: Recent case reports have described the efficacy of daratumumab to treat refractory pure red cell aplasia (PRCA) following major ABO mismatched allogeneic hematopoietic stem cell transplantation (HSCT). In this report, we describe the use of daratumumab as a first-line agent for treatment of delayed red blood cell (RBC) engraftment following a major ABO mismatched pediatric HSCT and provide a review of the literature. STUDY DESIGN AND MATERIALS: We report on a 14-year-old with DOCK8 deficiency who underwent a myeloablative, haploidentical bone marrow transplant from her major ABO mismatched sister (recipient O+, donor A+) for treatment of her primary immunodeficiency. Despite achieving full donor chimerism, she had delayed RBC engraftment requiring ongoing transfusions. Due to iron deposition, symptomatic anemia, and persistence of anti-A iso-hemagglutinins despite discontinuation of immunosuppression, treatment for delayed RBC engraftment with the CD38-targeted monoclonal antibody daratumumab was selected as a less immunosuppressive agent that could more selectively target iso-hemagglutinin producing plasma cells without causing broad B-cell aplasia. RESULTS: Clinical effect with daratumumab was demonstrated by reduced iso-hemagglutinin titer, increased reticulocytosis, normalization of her hemoglobin, and transfusion independence. In the 11-month follow-up period to date, no additional transfusions or immunosuppression have been necessary, despite persistence of low-level anti-A iso-hemagglutinin. CONCLUSION: Our experience suggests that daratumumab was an effective first-line therapy for delayed RBC engraftment and that earlier consideration for daratumumab in treatment of delayed RBC engraftment may be warranted.


Assuntos
Sistema ABO de Grupos Sanguíneos/imunologia , Anticorpos Monoclonais/farmacologia , Transplante de Medula Óssea/métodos , Função Retardada do Enxerto/tratamento farmacológico , Fatores de Troca do Nucleotídeo Guanina/deficiência , Doenças da Imunodeficiência Primária/terapia , Adolescente , Assistência ao Convalescente , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Quimerismo , Eritrócitos/imunologia , Feminino , Hemaglutininas/sangue , Hemaglutininas/efeitos dos fármacos , Humanos , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/genética , Aplasia Pura de Série Vermelha/tratamento farmacológico , Transplante Haploidêntico/efeitos adversos , Resultado do Tratamento
5.
Blood ; 125(18): 2753-8, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25691160

RESUMO

Ras-associated autoimmune leukoproliferative disorder (RALD) is a chronic, nonmalignant condition that presents with persistent monocytosis and is often associated with leukocytosis, lymphoproliferation, and autoimmune phenomena. RALD has clinical and laboratory features that overlap with those of juvenile myelomonocytic leukemia (JMML) and chronic myelomonocytic leukemia (CMML), including identical somatic mutations in KRAS or NRAS genes noted in peripheral blood mononuclear cells. Long-term follow-up of these patients suggests that RALD has an indolent clinical course whereas JMML is fatal if left untreated. Immunophenotyping peripheral blood from RALD patients shows characteristic circulating activated monocytes and polyclonal CD10(+) B cells. Distinguishing RALD from JMML and CMML has implications for clinical care and prognosis.


Assuntos
Doenças Autoimunes/genética , Genes ras , Leucemia Mielomonocítica Juvenil/genética , Leucocitose/genética , Adolescente , Adulto , Doenças Autoimunes/patologia , Criança , Pré-Escolar , Feminino , Humanos , Leucemia Mielomonocítica Juvenil/patologia , Leucocitose/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Adulto Jovem
6.
Blood ; 125(1): 56-70, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25359990

RESUMO

Germ-line GATA2 gene mutations, leading to haploinsufficiency, have been identified in patients with familial myelodysplastic syndrome/acute myeloid leukemia, monocytopenia and mycobacterial infections, Emberger syndrome, and dendritic cell, monocyte, B-, and NK-cell deficiency. GATA2 mutations have also been reported in a minority of patients with congenital neutropenia and aplastic anemia (AA). The bone marrow (BM) from patients with GATA2 deficiency is typically hypocellular, with varying degrees of dysplasia. Distinguishing GATA2 patients from those with AA is critical for selecting appropriate therapy. We compared the BM flow cytometric, morphologic, and cytogenetic features of 28 GATA2 patients with those of 32 patients being evaluated for idiopathic AA. The marrow of GATA2 patients had severely reduced monocytes, B cells, and NK cells; absent hematogones; and inverted CD4:CD8 ratios. Atypical megakaryocytes and abnormal cytogenetics were more common in GATA2 marrows. CD34(+) cells were comparably reduced in GATA2 and AA. Using these criteria, we prospectively identified 4 of 32 patients with suspected AA who had features suspicious for GATA2 mutations, later confirmed by DNA sequencing. Our results show that routine BM flow cytometry, morphology, and cytogenetics in patients who present with cytopenia(s) can identify patients for whom GATA2 sequencing is indicated.


Assuntos
Anemia Aplástica/diagnóstico , Doenças da Medula Óssea/diagnóstico , Fator de Transcrição GATA2/deficiência , Adulto , Anemia Aplástica/genética , Anemia Aplástica/metabolismo , Antígenos CD34/metabolismo , Medula Óssea/patologia , Doenças da Medula Óssea/genética , Estudos de Coortes , Citogenética , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação , Análise de Sequência de DNA , Adulto Jovem
7.
J Immunol ; 194(6): 2561-8, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681347

RESUMO

Several potent and broadly neutralizing Abs to HIV-1 have been isolated recently from peripheral blood B cells of infected individuals, based on prescreening of Ab activity in the serum. However, little is known regarding the cells that make the Abs that circulate in the blood. Accordingly, we investigated the most likely source, the bone marrow, of chronically HIV-1-infected individuals who were not receiving antiretroviral therapy. Increased frequencies of plasma cells, as well as B cell precursors, namely preB-I and preB-II, and decreased frequencies of mature B cells were observed in bone marrow aspirates of these individuals compared with HIV-negative counterparts. Increased frequencies of bone marrow plasma cells are consistent with known hallmarks of HIV-1 infection, namely hypergammaglobulinemia and increased frequencies of peripheral blood plasmablasts. Levels of HIV-1 envelope (Env)-binding and HIV-1-neutralizing Abs were measured in serum, and corresponding frequencies of Ab-secreting or Env-binding cells were measured in the blood (plasmablasts and memory B cells) and in the bone marrow (plasma cells). A strong correlation was observed between serum HIV-1-specific Abs and Env-specific bone marrow-derived plasma cells, but not circulating plasmablasts or memory B cells. These findings demonstrate that, despite HIV-1-induced phenotypic and functional B cell dysregulation in the peripheral blood and secondary lymphoid tissues, bone marrow plasma cells remain a primary source for circulating HIV-1-specific Abs in HIV-1-infected individuals.


Assuntos
Células da Medula Óssea/imunologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Plasmócitos/imunologia , Adulto , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Medula Óssea/imunologia , Medula Óssea/virologia , Células da Medula Óssea/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Anticorpos Anti-HIV/sangue , Infecções por HIV/sangue , Infecções por HIV/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Memória Imunológica/imunologia , Contagem de Linfócitos , Masculino , Plasmócitos/metabolismo , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/metabolismo , Adulto Jovem , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo
9.
Am J Clin Pathol ; 157(5): 709-717, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35512254

RESUMO

OBJECTIVES: Immunohistochemistry (IHC) is a useful method for mantle cell lymphoma (MCL) detection in the bone marrow (BM). However, recognition of the neoplastic B cells can be challenging, especially when there is low-level disease. METHODS: We examined BM from 105 patients with MCL. IHC was performed using cyclin D1/CD79a and PAX5/CD5 dual stains, which were compared with single stains that included CD20, CD79a, cyclin D1, and CD5 and with multiparameter flow cytometry (FC). RESULTS: Based on the FC data, the overall sensitivity of the dual IHC stains was 95.6%. Both dual IHC stains showed better efficacy for detecting MCL cells compared with the aggregated single stains (P = .012). While three cases were positive by FC analysis but negative for dual staining, four cases showed cells positive for cyclin D1/CD79a and PAX5/CD5 dual staining that were not detected by FC. Two of these latter cases were in patients with minimal or focal disease involvement. CONCLUSIONS: Cyclin D1/CD79a and PAX5/CD5 dual IHC staining is an efficient procedure for the detection of MCL in the marrow and is particularly helpful in low-level or focal involvement by MCL. This approach can be particularly useful when marrow aspirates are inadequate or unavailable.


Assuntos
Linfoma de Célula do Manto , Adulto , Medula Óssea/patologia , Ciclina D1 , Humanos , Imuno-Histoquímica , Linfoma de Célula do Manto/diagnóstico , Linfoma de Célula do Manto/patologia , Coloração e Rotulagem
10.
Blood Adv ; 5(23): 4807-4816, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34607348

RESUMO

Hairy cell leukemia variant (HCLv) responds poorly to purine analogue monotherapy. Rituximab concurrent with cladribine (CDAR) improves response rates, but long-term outcomes are unknown. We report final results of a phase 2 study of CDAR for patients with HCLv. Twenty patients with 0 to 1 prior courses of cladribine and/or rituximab, including 8 who were previously untreated, received cladribine 0.15 mg/kg on days 1 to 5 with 8 weekly rituximab doses of 375 mg/m2 beginning day 1. Patients received a second rituximab course ≥6 months after cladribine, if and when minimal residual disease (MRD) was detected in blood. The complete remission (CR) rate from CDAR was 95% (95% confidence interval, 75-100). Sixteen (80%) of 20 patients (95% confidence interval, 56-94) became MRD negative according to bone marrow at 6 months. The median duration of MRD-negative CR was 70.1 months, and 7 of 16 are still MRD negative up to 120 months. With a median follow-up of 69.7 months, 11 patients received delayed rituximab, and the 5-year progression-free survival (PFS) and overall survival (OS) were 63.3% and 73.9%, respectively. Five patients with TP53 mutations had shorter PFS (median, 36.4 months vs unreached; P = .0024) and OS (median, 52.4 months vs unreached; P = .032). MRD-negative CR at 6 months was significantly associated with longer PFS (unreached vs 17.4 months; P < .0001) and OS (unreached vs 38.2 months; P < .0001). Lack of MRD in blood at 6 months was also predictive of longer PFS and OS (P < .0001). After progression following CDAR, median OS was 29.7 months. CDAR is effective in HCLv, with better outcomes in patients who achieve MRD-negative CR. This trial is registered at www.clinicaltrials.gov as #NCT00923013.


Assuntos
Cladribina , Leucemia de Células Pilosas , Cladribina/uso terapêutico , Seguimentos , Humanos , Leucemia de Células Pilosas/tratamento farmacológico , Indução de Remissão , Rituximab
11.
JAMA Oncol ; 7(11): 1678-1685, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34529025

RESUMO

IMPORTANCE: High-risk smoldering myeloma has a 5-year risk of progression to symptomatic multiple myeloma of approximately 75%. Treatment with lenalidomide decreases the risk of progression; however, novel triplet regimens are superior, and earlier disease may be more treatment sensitive. OBJECTIVE: To evaluate the use of carfilzomib, lenalidomide, and dexamethasone (KRd) with lenalidomide maintenance therapy as early intervention in high-risk smoldering myeloma and to determine the rates of minimal residual disease (MRD)-negative complete response (CR). DESIGN, SETTING, AND PARTICIPANTS: In this single-arm, single-center, phase 2 nonrandomized controlled trial, responses were evaluated at every cycle during KRd treatment and every 3 cycles subsequently. Bone marrow biopsies and imaging were performed by cycle 8 and then annually. The study enrolled patients from May 29, 2012, to July 23, 2020, at the National Institutes of Health Clinical Center, a highly specialized tertiary cancer center. Patient key eligibility criteria included a diagnosis of high-risk smoldering myeloma based on the Mayo Clinic, Spanish, and/or Rajkumar, Mateos, and Landgren criteria. INTERVENTIONS: Patients received eight 4-week cycles of intravenous carfilzomib 36 mg/m2 (first 2 doses, 20 mg/m2), dexamethasone (20 mg, cycles 1-4; 10 mg, cycles 5-8 twice weekly), and lenalidomide 25 mg (days 1-21) followed by twenty-four 28-day cycles of maintenance lenalidomide 10 mg (days 1-21). Stem cell harvest and storage were optional. MAIN OUTCOMES AND MEASURES: The primary outcome was the MRD-negative CR rate. Key secondary outcomes included duration of MRD-negative CR and progression to multiple myeloma. RESULTS: A total of 54 patients (median age, 59 years [range, 40-79 years]; 30 men [55.6%]; and 2 Asian [3.7%], 15 Black [27.8%], 1 Hispanic [1.9%], and 36 White [66.7%] patients) were enrolled, with a median potential follow-up time of 31.9 months (range, 6.7-102.9 months). The MRD-negative CR rate was 70.4% (95% CI, 56.4%-82.0%), with a median sustained duration of 5.5 years (95% CI, 3.7 years to not estimable). The 8-year probability of being free from progression to multiple myeloma was 91.2% (95% CI, 67.4%-97.9%), and no deaths occurred. Nonhematologic grade 3 adverse events occurred in 21 patients (38.9%) and included thromboembolism, rash, and lung infection, with no grade 4 events. CONCLUSIONS AND RELEVANCE: Results of this phase 2 nonrandomized controlled trial suggest that treatment of high-risk smoldering myeloma with novel triplet regimens, such as KRd and lenalidomide maintenance therapy, may alter the natural history of smoldering myeloma by significantly delaying development of end-organ disease. Randomized clinical trials are needed to confirm this favorable benefit-to-risk profile. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01572480.


Assuntos
Mieloma Múltiplo , Mieloma Múltiplo Latente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Dexametasona , Feminino , Humanos , Lenalidomida , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/patologia , Oligopeptídeos
12.
J Clin Oncol ; 38(14): 1527-1538, 2020 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-32109194

RESUMO

PURPOSE: Single-agent purine analog, usually cladribine, has been the standard first-line therapy of hairy cell leukemia (HCL) for 30 years. High complete remission (CR) rates often include minimal residual disease (MRD), leading to relapse and repeated treatments. Rituximab can clear MRD, but long-term results are unknown and optimal timing of rituximab undefined. PATIENTS AND METHODS: Patients were randomly assigned to first-line cladribine 0.15 mg/kg intravenously days 1-5 with 8 weekly doses of rituximab 375 mg/m2 begun either day 1 (concurrent, CDAR) or ≥ 6 months later (delayed) after detection of MRD in blood. MRD tests included blood and bone marrow (BM) flow cytometry, and BM immunohistochemistry. RESULTS: Sixty-eight patients with purine analog-naïve classic HCL were randomly assigned 1:1 to concurrent versus delayed arms. At 6 months after CDAR versus cladribine monotherapy, CR rates were 100% versus 88% (P = .11), MRD-free CR rates 97% versus 24% (P < .0001, primary end point), and blood MRD-free rates 100% versus 50% (P < .0001), respectively. At 96 months median follow-up, 94% versus 12% remained MRD free. Compared with CDAR, delayed rituximab after cladribine achieved lower rate (67% of 21 evaluable patients; P = .0034) and durability (P = .0081, hazard radio favoring CDAR, 0.094) of MRD-free CR. Nevertheless, 12 patients in the delayed arm remained MRD free when restaged 6-104 (median, 78) months after last delayed rituximab treatment. Compared with cladribine monotherapy, CDAR led to brief grade 3/4 thrombocytopenia (59% v 9%; P < .0001) and platelet transfusions without bleeding (35% v 0%; P = .0002), but higher neutrophil (P = .017) and platelet (P = .0015) counts at 4 weeks. CONCLUSION: Achieving MRD-free CR of HCL after first-line cladribine is greatly enhanced by concurrent rituximab and less so by delayed rituximab. Longer follow-up will determine if MRD-free survival leads to less need for additional therapy or cure of HCL.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cladribina/uso terapêutico , Leucemia de Células Pilosas/tratamento farmacológico , Rituximab/uso terapêutico , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cladribina/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Rituximab/farmacologia
14.
J Pediatr Hematol Oncol ; 31(2): 145-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19194204

RESUMO

Anaplastic large cell lymphoma (ALCL) is a unique clinical and pathologic subtype of lymphoma characterized by the proliferation of large, highly pleomorphic CD30-positive cells. Overall 70% to 80% of children with ALCL are cured with modern chemotherapy regimens, but the disease is often resistant to multiple therapies after relapse. Single agent vinblastine therapy has been effective in some cases of refractory ALCL. We report a case of ALCL originally diagnosed in an 8-year-old girl. After relapse, the disease was refractory to multiagent chemotherapy, but has showed remarkable response to, and dependence on, single agent vinblastine treatment for almost 7 years.


Assuntos
Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Vimblastina/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Criança , Feminino , Humanos , Recidiva , Indução de Remissão/métodos , Terapia de Salvação
15.
Mol Cancer Ther ; 7(8): 2308-18, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18723478

RESUMO

Jak2 tyrosine kinase is essential for animal development and hyperkinetic Jak2 function has been linked to a host of human diseases. Control of this pathway using Jak2-specific inhibitors would therefore potentially serve as a useful research tool and/or therapeutic agent. Here, we used a high-throughput program called DOCK to predict the ability of 20,000 small molecules to interact with a structural pocket adjacent to the ATP-binding site of murine Jak2. One small molecule, 2-methyl-1-phenyl-4-pyridin-2-yl-2-(2-pyridin-2-ylethyl)butan-1-one (herein designated as Z3), bound to Jak2 with a favorable energy score. Z3 inhibited Jak2-V617F and Jak2-WT autophosphorylation in a dose-dependent manner but was not cytotoxic to cells at concentrations that inhibited kinase activity. Z3 selectively inhibited Jak2 kinase function with no effect on Tyk2 or c-Src kinase function. Z3 significantly inhibited proliferation of the Jak2-V617F-expressing, human erythroleukemia cell line, HEL 92.1.7. The Z3-mediated reduction in cell proliferation correlated with reduced Jak2 and STAT3 tyrosine phosphorylation levels as well as marked cell cycle arrest. Finally, Z3 inhibited the growth of hematopoietic progenitor cells isolated from the bone marrow of an essential thrombocythemia patient harboring the Jak2-V617F mutation and a polycythemia vera patient carrying a Jak2-F537I mutation. Collectively, the data suggest that Z3 is a novel specific inhibitor of Jak2 tyrosine kinase.


Assuntos
Butanonas/farmacologia , Divisão Celular/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Imunoprecipitação , Janus Quinase 2/metabolismo , Fosforilação
16.
Methods Mol Biol ; 2032: 281-296, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31522424

RESUMO

Immunophenotyping by multiparameter flow cytometry is a rapid and efficient technique to simultaneously assess and correlate multiple individual cell properties like size and internal complexity along with antigen expression in a population of cells. This method is utilized for rapid characterization of the blasts and classification of acute myeloid leukemia (AML), in both the peripheral blood (PB) and bone marrow (BM). This technique is not only useful in the initial diagnosis but also in monitoring and determining prognosis of the disease through minimal residual disease (MRD) testing. This chapter provides an overview of procedures for specimen processing, staining, and immunophenotyping of AML and describes the principles of data analysis for AML classification and MRD testing.


Assuntos
Citometria de Fluxo/métodos , Imunofenotipagem/métodos , Leucemia Mieloide Aguda/sangue , Antígenos/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Leucemia Mieloide Aguda/imunologia
17.
Semin Hematol ; 56(1): 58-64, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573046

RESUMO

Enumeration of blasts in the bone marrow is critical for diagnostic, prognostic, and therapeutic response evaluation in myelodysplastic syndromes, myeloproliferative neoplasms and acute leukemias. However, few studies have examined the accuracy and precision of marrow blast counting using standard microscopic procedures. In our study, 4 experienced hematopathologists evaluated blast percentages in marrow using either differential counts on aspirate smears or visual estimates on CD34-stained trephine biopsies. Results of an independent observer's manual counts of individual labeled and unlabeled cells performed on high resolution digital images of CD34-stained trephine biopsies were designated as the "Digital Reference." Hematopathologists' blast counts showed excellent interobserver reproducibility, but the counts in smears and trephine biopsies correlated poorly with each other. Compared to the Digital Reference, both smear and trephine evaluations showed positive bias and high variability. The biopsy showed less variability but higher positive bias relative to the smears, indicating that counts were overestimated more in the hematopathologists' biopsy evaluation. Flow cytometric counts correlated well with the Digital Reference, and cases with high blast count generally showed worse cytogenetic findings. Our results demonstrate the need for better counting methods if significant decisions are made based on microscopic enumeration of blasts. Further efforts should be made to develop markers to better define blast cells and perhaps incorporate automated digital imaging technologies to enumerate them. Also, consideration should be given to quantifying blasts per marrow area in biopsies instead of per nucleated cells.


Assuntos
Células da Medula Óssea/metabolismo , Medula Óssea/patologia , Leucemia/patologia , Síndromes Mielodisplásicas/patologia , Transtornos Mieloproliferativos/patologia , Células da Medula Óssea/citologia , Humanos , Prognóstico
18.
Semin Hematol ; 56(1): 65-68, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573047

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired condition in which, due to a mutation of the phosphatidylinositol glycan class A gene, hematopoietic cells lack proteins that are normally anchored to the cell surface by glycosylphosphatidylinositol (GPI). Thus, PNH cells show poor expression of surface proteins, such as CD55 and CD59, and dim or absent binding of fluorescently labeled modified aerolysin (FLAER). In clinical diagnostic laboratories, the detection and quantitation of PNH is currently performed by flow cytometry (FC) analysis of peripheral blood (PB) samples. Although PB remains the preferred source of cells for PNH detection, we and other authors have shown that a careful FC analysis of bone marrow (BM) aspirates can provide results for PNH detection and quantitation equivalent to those obtained with PB. Here, we review studies delineating the expression of GPI-anchored proteins and FLAER binding in normal BM cells, and summarize published findings demonstrating the feasibility of identifying and quantitating PNH cells in BM using FLAER as well as antibodies against GPI-anchored proteins. Detection of PNH cells in BM should be useful in patients with unsuspected PNH or with severe cytopenia in whom PB FC analysis may be challenging.


Assuntos
Hemoglobinúria Paroxística/diagnóstico , Medula Óssea , Citometria de Fluxo , Hemoglobinúria Paroxística/patologia , Humanos
19.
Lab Invest ; 88(12): 1258-63, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19020521

RESUMO

The close of a 5-year editorship gives opportunity to reflect on the highs and the lows of an editorship. The goal of such reflection is to assist both authors and reviewers in interacting with a biomedical journal, and to foster interest among individuals contemplating an editorship. Among the highs was the privilege of publishing high-quality original scientific work within the scope of the journal; in this instance mechanistic studies of disease. Although review articles and editorials have their reward, it is the publication of original peer-reviewed work that constitutes the true basis for advancing biomedical science. This is the heart of journal publication. Second, the editorial interaction with submitting authors in bringing their work to publication is itself highly rewarding, and can lead to longer-term collegial working relationships between editors and authors. The anonymous expert reviewers also play a key role in bringing outstanding scientific work to successful publication. Collectively, authors, editors, and reviewers constitute an important 'community of science'. Third, working together as an editorial team, especially through the weekly 'journal clubs' that a regular editorial meeting affords, is commended as a key reward of any editorial group taking on journal management. The lows included sifting through submitted manuscripts in which the rigor of science was not satisfactory, and encountering specific instances of compromised scientific integrity--fortunately rare. In both instances, the editorial commitment is to publish high-quality original science; a necessary corollary is identifying those submissions, through rigorous but fair review, which do not meet journal standards. In the end, editorship is a highly rewarding experience, and very much conducive to sustaining the wonder of science that drew us to this profession.


Assuntos
Jornalismo Médico , Editoração , Revisão da Pesquisa por Pares
20.
Pediatr Blood Cancer ; 50(2): 415-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17051534

RESUMO

Post-transplant lymphoproliferative disorder (PTLD) is the most common treatment related malignancy that occurs after solid organ transplantation (SOT). PTLD has extended from its initial description as an Epstein-Barr virus (EBV)-driven B-cell proliferation to include EBV-negative and non B-lineage cases. T-cell PTLD (T-PTLD) is rare in both adults and children. We report two cases of pediatric T-PTLD after SOT (liver and lungs) and review cases reported in the literature. Both patients had a bimodal response to therapy with initial eradication of bulky nodal disease with regimens typically used to treat leukemia, but persistence of low-level clonal T-cells in marrow, CSF and lung in one case.


Assuntos
Transplante de Fígado/efeitos adversos , Transplante de Pulmão/efeitos adversos , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/patologia , Linfócitos T/patologia , Pré-Escolar , Feminino , Humanos , Lactente
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa