Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Blood ; 143(21): 2190-2200, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38306657

RESUMEN

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.


Asunto(s)
Trombosis , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Trombosis/etiología , Trombosis/genética , Trombosis/epidemiología , Adolescente , Enzimas Activadoras de Ubiquitina/genética , Adulto Joven , Factores de Riesgo , Anciano , Niño , Trombosis de la Vena/etiología , Trombosis de la Vena/epidemiología , Trombosis de la Vena/genética , Incidencia , Mutación , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/complicaciones , Preescolar
3.
Blood Adv ; 5(23): 4807-4816, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34607348

RESUMEN

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.


Asunto(s)
Cladribina , Leucemia de Células Pilosas , Cladribina/uso terapéutico , Estudios de Seguimiento , Humanos , Leucemia de Células Pilosas/tratamiento farmacológico , Inducción de Remisión , Rituximab
4.
J Clin Oncol ; 38(14): 1527-1538, 2020 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-32109194

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cladribina/uso terapéutico , Leucemia de Células Pilosas/tratamiento farmacológico , Rituximab/uso terapéutico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cladribina/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Rituximab/farmacología
5.
Semin Hematol ; 56(1): 1, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573038
6.
Semin Hematol ; 56(1): 65-68, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30573047

RESUMEN

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.


Asunto(s)
Hemoglobinuria Paroxística/diagnóstico , Médula Ósea , Citometría de Flujo , Hemoglobinuria Paroxística/patología , Humanos
7.
Am J Clin Pathol ; 150(3): 273-282, 2018 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-29982419

RESUMEN

OBJECTIVES: To determine fluorescently labeled aerolysin (FLAER) binding and glycophosphatidylinositol-anchored protein expression in bone marrow (BM) cells of healthy volunteers and patients with paroxysmal nocturnal hemoglobinuria (PNH) detected in peripheral blood (PB); compare PNH clone size in BM and PB; and detect PNH in BM by commonly used antibodies. METHODS: Flow cytometry analysis of FLAER binding to leukocytes and expression of CD55/CD59 in erythrocytes. Analysis of CD16 in neutrophils and CD14 in monocytes in BM. RESULTS: FLAER binds to all normal BM leukocytes, and binding increases with cell maturation. In PNH, lymphocytic clones are consistently smaller than clones of other BM cells. PNH clones are detectable in mature BM leukocytes with high specificity and sensitivity using common antibodies. CONCLUSIONS: PNH clone sizes measured in mature BM leukocytes and in PB are comparable, making BM suitable for PNH assessment. We further demonstrate that commonly used reagents (not FLAER or CD55/CD59) can reliably identify abnormalities of BM neutrophils and monocytes consistent with PNH cells.

9.
Clin Cancer Res ; 22(7): 1572-82, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26660519

RESUMEN

PURPOSE: Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental interactions for proliferation and survival that are at least partially mediated through B-cell receptor (BCR) signaling. Ibrutinib, a Bruton tyrosine kinase inhibitor, disrupts BCR signaling and leads to the egress of tumor cells from the microenvironment. Although the on-target effects on CLL cells are well defined, the impact on the microenvironment is less well studied. We therefore sought to characterize the in vivo effects of ibrutinib on the tumor microenvironment. EXPERIMENTAL DESIGN: Patients received single-agent ibrutinib on an investigator-initiated phase II trial. Serial blood and tissue samples were collected pretreatment and during treatment. Changes in cytokine levels, cellular subsets, and microenvironmental interactions were assessed. RESULTS: Serum levels of key chemokines and inflammatory cytokines decreased significantly in patients on ibrutinib. Furthermore, ibrutinib treatment decreased circulating tumor cells and overall T-cell numbers. Most notably, a reduced frequency of the Th17 subset of CD4(+)T cells was observed concurrent with reduced expression of activation markers and PD-1 on T cells. Consistent with direct inhibition of T cells, ibrutinib inhibited Th17 differentiation of murine CD4(+)T cells in vitro Finally, in the bone marrow microenvironment, we found that ibrutinib disaggregated the interactions of macrophages and CLL cells, inhibited secretion of CXCL13, and decreased the chemoattraction of CLL cells. CONCLUSIONS: In conjunction with inhibition of BCR signaling, these changes in the tumor microenvironment likely contribute to the antitumor activity of ibrutinib and may impact the efficacy of immunotherapeutic strategies in patients with CLL. See related commentary by Bachireddy and Wu, p. 1547.


Asunto(s)
Antineoplásicos/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Microambiente Tumoral/efectos de los fármacos , Adenina/análogos & derivados , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Médula Ósea/patología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/inmunología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunofenotipificación , Mediadores de Inflamación/sangre , Mediadores de Inflamación/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Piperidinas , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo
10.
J Immunol ; 194(6): 2561-8, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25681347

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/inmunología , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Células Plasmáticas/inmunología , Adulto , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Médula Ósea/inmunología , Médula Ósea/virología , Células de la Médula Ósea/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/sangre , Infecciones por VIH/virología , VIH-1/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Memoria Inmunológica/inmunología , Recuento de Linfocitos , Masculino , Células Plasmáticas/metabolismo , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Adulto Joven , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
11.
Blood ; 125(18): 2753-8, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25691160

RESUMEN

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.


Asunto(s)
Enfermedades Autoinmunes/genética , Genes ras , Leucemia Mielomonocítica Juvenil/genética , Leucocitosis/genética , Adolescente , Adulto , Enfermedades Autoinmunes/patología , Niño , Preescolar , Femenino , Humanos , Leucemia Mielomonocítica Juvenil/patología , Leucocitosis/patología , Masculino , Persona de Mediana Edad , Mutación , Adulto Joven
12.
Blood ; 125(1): 56-70, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25359990

RESUMEN

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.


Asunto(s)
Anemia Aplásica/diagnóstico , Enfermedades de la Médula Ósea/diagnóstico , Factor de Transcripción GATA2/deficiencia , Adulto , Anemia Aplásica/genética , Anemia Aplásica/metabolismo , Antígenos CD34/metabolismo , Médula Ósea/patología , Enfermedades de la Médula Ósea/genética , Estudios de Cohortes , Citogenética , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mutación , Análisis de Secuencia de ADN , Adulto Joven
13.
Int J Clin Exp Pathol ; 5(2): 110-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22400070

RESUMEN

The diagnosis of B-cell lymphoma (BCL) is often dependent on the detection of clonal immunoglobulin (Ig) light chain expression. In some BCLs, the determination of clonality based on Ig light chain restriction may be difficult. The aim of our study was to assess the utility of flow cytometric analysis of surface Ig heavy chain (HC) expression in lymphoid tissues in distinguishing lymphoid hyperplasias from BCLs, and also differentiating various BCL subtypes. HC expression on B-cells varied among different types of hyperplasias. In follicular hyperplasia, IgM and IgD expression was high in mantle cells while germinal center cells showed poor HC expression. In other hyperplasias, B cell compartments were blurred but generally showed high IgD and IgM expression. Compared to hyperplasias, BCLs varied in IgM expression. Small lymphocytic lymphomas had lower IgM expression than mantle cell lymphomas. Of importance, IgD expression was significantly lower in BCLs than in hyperplasias, a finding that can be useful in differentiating lymphoma from reactive processes.


Asunto(s)
Citometría de Flujo/métodos , Cadenas Pesadas de Inmunoglobulina/inmunología , Linfoma de Células B/diagnóstico , Seudolinfoma/diagnóstico , Diagnóstico Diferencial , Expresión Génica , Perfilación de la Expresión Génica , Centro Germinal/inmunología , Centro Germinal/patología , Humanos , Inmunoglobulina D/genética , Inmunoglobulina D/inmunología , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Inmunofenotipificación , Linfoma de Células B/inmunología , Seudolinfoma/inmunología
14.
Mol Cancer Ther ; 7(8): 2308-18, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723478

RESUMEN

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.


Asunto(s)
Butanonas/farmacología , División Celular/efectos de los fármacos , Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Adenosina Trifosfato/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Inmunoprecipitación , Janus Quinasa 2/metabolismo , Fosforilación
15.
Hum Pathol ; 38(12): 1779-87, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17714757

RESUMEN

Assessment of bone marrow involvement by malignant plasma cells is an important element in the diagnosis and follow-up of patients with multiple myeloma and other plasma cell dyscrasias. Microscope-based differential counts of bone marrow aspirates are used as the primary method to evaluate bone marrow plasma cell percentages. However, multiple myeloma is often a focal process, a fact that impacts the accuracy and reliability of the results of bone marrow plasma cell percentages obtained by differential counts of bone marrow aspirate smears. Moreover, the interobserver and intraobserver reproducibility of counting bone marrow plasma cells microscopically has not been adequately tested. CD138 allows excellent assessment of plasma cell numbers and distribution in bone marrow biopsies. We compared estimates of plasma cell percentages in bone marrow aspirates and in hematoxylin-eosin- and CD138-stained bone marrow biopsy sections (CD138 sections) in 79 bone marrows from patients with multiple myeloma. There was a notable discrepancy in bone marrow plasma cell percentages using the different methods of observation. In particular, there was a relatively poor concordance of plasma cell percentage estimation between aspirate smears and CD138 sections. Estimates of plasma cell percentage using CD138 sections demonstrated the highest interobserver concordance. This observation was supported by computer-assisted image analysis. In addition, CD138 expression highlighted patterns of plasma cell infiltration indicative of neoplasia even in the absence of plasmacytosis. We conclude that examination of CD138 sections should be considered for routine use in the estimation of plasma cell load in the bone marrow.


Asunto(s)
Células de la Médula Ósea/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/metabolismo , Sindecano-1/biosíntesis , Células de la Médula Ósea/patología , Examen de la Médula Ósea , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Variaciones Dependientes del Observador , Células Plasmáticas/patología , Pronóstico
16.
J Nucl Med ; 48(4): 645-54, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17401104

RESUMEN

UNLABELLED: Current bone marrow dosimetry methods inherently assume that the target cells of interest for the assessment of leukemia risk (stochastic effects) or marrow toxicity (deterministic effects) are uniformly localized throughout the marrow cavities of cancellous bone. Previous studies on mouse femur, however, have demonstrated a spatial gradient for the hematopoietic stem and progenitor cells, with higher concentrations near the bone surfaces. The objective of the present study was to directly measure the spatial concentration of these cells, as well as marrow vasculature structures, within images of human disease-free bone marrow. METHODS: Core-biopsy samples of normal bone marrow from the iliac crest were obtained from clinical cases at Shands Hospital at the University of Florida Department of Pathology. The specimens were sectioned and immunohistochemically stained for CD34 (red) and CD31 (brown) antigens. These 2 stains were used simultaneously to differentiate between hematopoietic stem and progenitor cells (CD34(+)/CD31(-)) and vascular endothelium (CD34(+)/CD31(+)). Distances from hematopoietic CD34(+) cells and blood vessels to the nearest bone trabecula surface were measured digitally and then binned in 50-mum increments, with the results then normalized per unit area of marrow tissue. The distances separating hematopoietic CD34(+) cells from vessels were also tallied. RESULTS: Hematopoietic CD34(+) cells were found to exist along a linear spatial gradient with a maximal areal concentration localized within the first 50 mum of the bone surfaces. An exponential spatial concentration gradient was found in the concentration of blood vessel fragments within the images. Distances between hematopoietic CD34(+) cells and blood vessels exhibited a lognormal distribution indicating a shared spatial niche. CONCLUSION: Study results confirm that the spatial gradient of hematopoietic stem and progenitor cells previously measured in mouse femur is also present within human cancellous bone. The dosimetric implication of these results may be significant for those scenarios in which the absorbed dose itself is nonuniformly delivered across the marrow tissues, as would be the case for a low-energy beta- or alpha-particle emitter localized on the bone surfaces.


Asunto(s)
Antígenos CD34/biosíntesis , Huesos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre/citología , Adulto , Anciano , Anciano de 80 o más Años , Huesos/efectos de la radiación , Niño , Preescolar , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis
17.
Clin Lymphoma Myeloma ; 7(4): 319-22, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17324342

RESUMEN

Immunohistochemistry using the L26 antibody recognizes an intracellular domain of CD20, whereas the L27 antibody used for surface CD20 staining by flow cytometry (FC) recognizes an extracellular domain and would be expected to be a better predictor of response to rituximab. We present a 75-year-old man who was initially treated for CD20- diffuse large B-cell lymphoma based on FC and, at relapse, still had CD20- disease by FC but CD20+ disease by immunohistochemistry. The patient responded to rituximab alone. On further study, it was shown that the malignant B cells, but not normal B cells, expressed the L27 surface binding site only within the intracellular domain. Therefore, it appears that the rituximab binding site is distinct from the surface binding site, and when there is a disparity between the methods to detect CD20 expression, consideration should be given to include rituximab in the treatment plan.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/biosíntesis , Citometría de Flujo/métodos , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales de Origen Murino , Antígenos CD20/inmunología , Antígenos CD20/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Sitios de Unión , Ciclofosfamida/uso terapéutico , Doxorrubicina/uso terapéutico , Resultado Fatal , Humanos , Inmunohistoquímica , Inmunofenotipificación , Masculino , Valor Predictivo de las Pruebas , Prednisona/uso terapéutico , Recurrencia , Rituximab , Sensibilidad y Especificidad , Tomografía Computarizada de Emisión/métodos , Resultado del Tratamiento , Vincristina/uso terapéutico
18.
Am J Clin Pathol ; 123(1): 119-24, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15762287

RESUMEN

We used flow cytometry to determine the CD58 expression on nonmalignant B cells at different stages of maturation in the bone marrow and compared it with that of blasts in adult and pediatric precursor B-cell acute lymphoblastic leukemia (B-ALL). The mean fluorescence intensity (MFI) of CD58 expression decreased significantly as nonmalignant B cells differentiated in the bone marrow from an early to a mature stage. Few nonneoplastic B cells at a mid or mature stage of development expressed CD58 MFI values comparable to those seen in leukemic cases. Early-stage nonneoplastic B-cell precursors expressed relatively higher CD58 levels, which frequently overlapped with the variable level of CD58 expression observed among leukemic blasts. As a group, however, the malignant precursor B-ALL cells showed significantly higher expression of CD58 than nonmalignant B-cell populations at any maturational stage. These findings support the potential usefulness of CD58 expression in the diagnosis and monitoring of precursor B-ALL, but only when blasts express high levels of CD58.


Asunto(s)
Linfocitos B/fisiología , Células de la Médula Ósea/fisiología , Antígenos CD58/análisis , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Adolescente , Adulto , Anciano , Antígenos CD34/análisis , Niño , Preescolar , Humanos , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...