Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hematol ; 96(5): 552-560, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33650179

RESUMEN

The MCRN-003/CCTGMYX.1 is a single arm phase II trial of weekly carfilzomib, cyclophosphamide and dexamethasone (wKCd), exploring a convenient immunomodulator (IMiD)-free regimen in relapsed myeloma. Weekly carfilzomib (20/70 mg/m2 ), dexamethasone 40 mg and cyclophosphamide 300 mg/m2 was delivered over 28-day cycles. The primary endpoint was overall response after four cycles. Secondary endpoints included toxicity, response depth, PFS and OS. Exploratory endpoints included the impact of cytogenetics, prior therapy exposure and serum free light chain (sFLC) escape; 76 patients were accrued. The ORR was 85% (68% ≥very good partial response [VGPR] and 29% ≥complete response [CR]). The median OS and PFS were 27 and 17 months respectively. High-risk cytogenetics conferred a worse ORR (75% vs. 97%, p = .013) and median OS (18 months vs. NR, p = .002) with a trend toward a worse median PFS (14 vs. 22 months, p = .06). Prior proteasome inhibitor (PI) or lenalidomide did not influence OS or PFS. The sFLC was noted in 15% of patients with a median PFS of 17 months when included as a progression event. The most common ≥ grade 3 non-hematologic adverse events were infectious (40%), vascular (17%) and cardiac (15%). The wKCD is a safe and effective regimen in relapse, especially for patients ineligible for lenalidomide-based therapies.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Enfermedades Cardiovasculares/inducido químicamente , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Dexametasona/administración & dosificación , Dexametasona/efectos adversos , Esquema de Medicación , Disnea/inducido químicamente , Femenino , Enfermedades Hematológicas/inducido químicamente , Humanos , Infecciones/etiología , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/genética , Proteínas de Mieloma/análisis , Oligopéptidos/administración & dosificación , Oligopéptidos/efectos adversos , Selección de Paciente , Pronóstico , Supervivencia sin Progresión , Recurrencia , Terapia Recuperativa , Resultado del Tratamiento
2.
N Engl J Med ; 371(10): 906-17, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25184863

RESUMEN

BACKGROUND: The combination melphalan-prednisone-thalidomide (MPT) is considered a standard therapy for patients with myeloma who are ineligible for stem-cell transplantation. However, emerging data on the use of lenalidomide and low-dose dexamethasone warrant a prospective comparison of the two approaches. METHODS: We randomly assigned 1623 patients to lenalidomide and dexamethasone in 28-day cycles until disease progression (535 patients), to the same combination for 72 weeks (18 cycles; 541 patients), or to MPT for 72 weeks (547 patients). The primary end point was progression-free survival with continuous lenalidomide-dexamethasone versus MPT. RESULTS: The median progression-free survival was 25.5 months with continuous lenalidomide-dexamethasone, 20.7 months with 18 cycles of lenalidomide-dexamethasone, and 21.2 months with MPT (hazard ratio for the risk of progression or death, 0.72 for continuous lenalidomide-dexamethasone vs. MPT and 0.70 for continuous lenalidomide-dexamethasone vs. 18 cycles of lenalidomide-dexamethasone; P<0.001 for both comparisons). Continuous lenalidomide-dexamethasone was superior to MPT for all secondary efficacy end points, including overall survival (at the interim analysis). Overall survival at 4 years was 59% with continuous lenalidomide-dexamethasone, 56% with 18 cycles of lenalidomide-dexamethasone, and 51% with MPT. Grade 3 or 4 adverse events were somewhat less frequent with continuous lenalidomide-dexamethasone than with MPT (70% vs. 78%). As compared with MPT, continuous lenalidomide-dexamethasone was associated with fewer hematologic and neurologic toxic events, a moderate increase in infections, and fewer second primary hematologic cancers. CONCLUSIONS: As compared with MPT, continuous lenalidomide-dexamethasone given until disease progression was associated with a significant improvement in progression-free survival, with an overall survival benefit at the interim analysis, among patients with newly diagnosed multiple myeloma who were ineligible for stem-cell transplantation. (Funded by Intergroupe, Francophone du Myélome and Celgene; FIRST ClinicalTrials.gov number, NCT00689936; European Union Drug Regulating Authorities Clinical Trials number, 2007-004823-39.).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Dexametasona/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Dexametasona/efectos adversos , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Lenalidomida , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Mieloma Múltiple/inmunología , Prednisona/administración & dosificación , Talidomida/administración & dosificación , Talidomida/efectos adversos
3.
Immunol Cell Biol ; 95(7): 611-619, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28303902

RESUMEN

MS4A4A is a member of the membrane-spanning, four domain family, subfamily A (MS4A) that includes CD20 (MS4A1), FcRß (MS4A2) and Htm4 (MS4A3). Like the first three members of this family, transcription of MS4A4A appears to be limited to hematopoietic cells. To evaluate expression of the MS4A4A protein in hematopoietic cell lineages and subsets we generated monoclonal antibodies against extracellular epitopes for use in flow cytometry. In human peripheral blood we found that MS4A4A is expressed at the plasma membrane in monocytes but not in granulocytes or lymphocytes. In vitro differentiation of monocytes demonstrated that MS4A4A is expressed in immature but not activated dendritic cells, and in macrophages generated in the presence of interleukin-4 ('alternatively activated' or M2 macrophages) but not by interferon-γ and lipopolysaccharide ('classically' activated or M1 macrophages). MS4A4A was expressed in the U937 monocytic cell line only after differentiation. In normal bone marrow, MS4A4A was expressed in mature monocytes but was undetected, or detected at only a low level, in myeloid/monocytic precursors, as well as their malignant counterparts in patients with various subtypes of myeloid leukemia. Although MS4A4A was not expressed in healthy B lymphocytes, it was highly expressed in normal plasma cells, CD138+ cells from multiple myeloma patients, and bone marrow B cells from a patient with mantle cell lymphoma. These findings suggest immunotherapeutic potential for MS4A4A antibodies in targeting alternatively activated macrophages such as tumor-associated macrophages, and in the treatment of multiple myeloma and mantle cell lymphoma.


Asunto(s)
Membrana Celular/metabolismo , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Células Plasmáticas/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Biomarcadores/metabolismo , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Leucemia Mieloide/inmunología , Leucemia Mieloide/patología , Macrófagos/efectos de los fármacos , Proteínas de la Membrana/sangre , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Células Plasmáticas/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Células U937 , Regulación hacia Arriba/efectos de los fármacos
4.
Cell Biol Toxicol ; 33(2): 83-97, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27761761

RESUMEN

Cancer heterogeneity is a significant factor in response to treatment and escape leading to relapse. Within an individual cancer, especially blood cancers, there exists multiple subclones as well as distinct clonal expansions unrelated to the clinically detected, dominant clone. Over time, multiple subclones and clones undergo emergence, expansion, and extinction. Although sometimes this intra-clonal and inter-clonal heterogeneity can be detected and/or quantified in tests that measure aggregate populations of cells, frequently, such heterogeneity can only be detected using single cell analysis to determine its frequency and to detect minor clones that may subsequently emerge to become drug resistant and dominant. Most genetic/genomic tests look at the pooled tumor population as a whole rather than at its individual cellular components. Yet, minor clones and cancer stem cells are unlikely to be detected against the background of expanded major clones. Because selective pressures are likely to govern much of what is seen clinically, single cell analysis allows identification of otherwise cryptic compartments of the malignancy that may ultimately mediate progression and relapse. Single cell analysis can track intra- or inter-clonal heterogeneity and provide useful clinical information, often before changes in the disease are detectable in the clinic. To a very limited extent, single cell analysis has already found roles in clinical care. Because inter- and intra-clonal heterogeneity likely occurs more frequently than can be currently appreciated on a clinical level, future use of single cell analysis is likely to have profound clinical utility.


Asunto(s)
Neoplasias Hematológicas/patología , Análisis de la Célula Individual/métodos , Animales , Toma de Decisiones Clínicas , Células Clonales , Neoplasias Hematológicas/terapia , Humanos
5.
Genes Chromosomes Cancer ; 55(12): 962-974, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27509849

RESUMEN

Many B-cell malignancies are characterized by chromosomal translocations involving IGH and a proto-oncogene. For translocations to occur, spatial proximity of translocation-prone genes is necessary. Currently, it is not known how such genes are brought into proximity with one another. Although decondensed chromosomes occupy definitive, non-random spaces in the interphase nucleus known as chromosome territories (CTs), chromatin at the edges of CTs can intermingle, and specific genomic regions from some chromosomes have been shown to "loop out" of their respective CTs. This extra-territorial positioning of specific genomic regions may provide a mechanism whereby translocation-prone genes are brought together in the interphase nucleus. FGFR3 and MAF recurrently participate in translocations with IGH at different frequencies. Using 3D, 4-color FISH, and 3D analysis software, we show frequent extra-territorial positioning of FGFR3 and significantly less frequent extra-territorial positioning of MAF. Frequent extra-territorial positioning may be characteristic of FGFR3 in B-cells from healthy adult donors and non-malignant B-cells from patients, but not in hematopoietic stem cells from patients with translocations. The frequency of extra-territorial positioning of FGFR3 and MAF in B-cells correlates with the frequency of translocations in the patient population. Most importantly, in patient B-cells, we demonstrate a significant proportion of extra-territorial FGFR3 participating in close loci pairs and/or colocalizing with IGH. This preliminary work suggests that in patient B-cells, extra-territorial positioning of FGFR3 may provide a mechanism for forming close loci pairs and/or colocalization with IGH; indirectly facilitating translocation events involving these two genes. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Núcleo Celular/genética , Cadenas Pesadas de Inmunoglobulina/genética , Interfase/genética , Mieloma Múltiple/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Represoras/genética , Adulto , Linfocitos B , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 4/genética , Femenino , Estudios de Seguimiento , Humanos , Hibridación Fluorescente in Situ , Masculino , Mieloma Múltiple/patología , Proto-Oncogenes Mas , Translocación Genética
6.
Genes Chromosomes Cancer ; 52(6): 523-37, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23460268

RESUMEN

Gene organization in nonmalignant B cells from t(4;14) and t(11;14) multiple myeloma (MM) patients differs from that of healthy donors. Among recurrent IGH translocations in MM, the frequency of t(4;14) (IGH and FGFR3) or t(11;14) (IGH and CCND1) is greater than the frequency of t(14;16) (IGH and MAF). Gene organization in t(14;16) patients may influence translocation potential of MAF with IGH. In patients, three-dimensional FISH revealed the positions of IGH, CCND1, FGFR3, and MAF in nonmalignant B cells that are likely similar to those when MM first arose, compared with B cells from healthy donors. Overall, IGH occupies a more central nuclear position while MAF is more peripherally located. However, for B cells from t(4;14) and t(11;14) patients, IGH and FGFR3, or IGH and CCND1 are found in spatial proximity: IGH and MAF are not. This differs in B cells from t(14;16) patients and healthy donors where IGH is approximately equidistant to FGFR3, CCND1, and MAF, suggesting that gene organization in t(14;16) patients is different from that in t(4;14) or t(11;14) patients. Translocations between IGH and MAF may arise only in the absence of close proximity to the more frequent partners, as appears to be the case for individuals who develop t(14;16) MM.


Asunto(s)
Linfocitos B/patología , Biomarcadores de Tumor/genética , Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 4/genética , Mieloma Múltiple/genética , Translocación Genética/genética , Linfocitos B/metabolismo , Sitios Genéticos , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Hibridación Fluorescente in Situ , Proteínas de Fusión Oncogénica/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-maf/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Genes Chromosomes Cancer ; 51(8): 727-42, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22489023

RESUMEN

Accumulating evidence suggests that spatial proximity of potential chromosomal translocation partners influences translocation probability. It is not known, however, whether genome organization differs in nonmalignant cells from patients as compared to their cellular counterparts from healthy donors. This could contribute to translocation potential causing cancer. Multiple myeloma is a hematopoietic cancer of the B-lineage, characterized by karyotypic instability, including chromosomal translocations involving the IGH locus and several translocation partners. Utilizing 3-D FISH and confocal imaging, we investigate whether nuclear spatial positioning of the translocation-prone gene loci, IGH, FGFR3, and CCND1 differs in nonmalignant cell subsets from multiple myeloma patients as compared to positioning in their corresponding healthy donor cell subsets. 3-D analysis software was used to determine the spatial proximity of potential translocation pairs and the radial distribution of each gene. We observed that in all cell subsets, the translocation-prone gene loci are intermediately located in the nucleus, while a control locus occupies a more peripheral position. In nonmalignant B-cells from multiple myeloma patients, however, the translocation-prone gene loci display a more central nuclear position and close spatial proximity. Our results demonstrate that gene positioning in nonmalignant B-cells from multiple myeloma patients differs from that in healthy donors, potentially contributing to translocation probability in patient cells. We speculate that genome reorganization in patient B-cells may closely reflect gene positioning at the time the multiple myeloma-specific translocation initially formed, thus influencing translocation probability between proximal loci in the B-cell population from which the malignancy emerged.


Asunto(s)
Linfocitos B/citología , Sitios Genéticos , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Translocación Genética , Células de la Médula Ósea/citología , Estudios de Casos y Controles , Núcleo Celular/genética , Ciclina D1/genética , ADN Intergénico , Células Madre Hematopoyéticas/patología , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Hibridación Fluorescente in Situ , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética
8.
Mol Cancer ; 11: 78, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23083101

RESUMEN

BACKGROUND: In multiple myeloma (MM), the immunoglobulin heavy chain VDJ gene rearrangement is a unique clonotypic signature that identifies all members of the myeloma clone independent of morphology or phenotype. Each clonotypic MM cell has only one genomic copy of the rearranged IgH VDJ. METHODS: Pre-treatment bone marrow aspirates from myeloma patients at diagnosis or in relapse were evaluated for the number of clonotypic cells using real time quantitative PCR (RPCR). RPCR measured the level of clonal cells, termed VDJ%, in 139 diagnosis and relapse BM aspirates from MM patients. RESULTS: Patients with a VDJ% below the median had a significantly longer event free survival (EFS) then those with a VDJ% higher than the median (p=0.0077, HR=0.57). Further, although the VDJ% from non-transplant patients predicted EFS (p=0.0093), VDJ% failed to predict outcome after autologous stem cell transplant (p=0.53). CONCLUSIONS: Our results suggest that for non-transplant patients, the tumor burden before treatment, perhaps reflecting cancer stem cell progeny/output, is an indirect measure that may indicate the number of MM cancer stem cells and hence event free survival.


Asunto(s)
Cadenas Pesadas de Inmunoglobulina/genética , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Recombinación V(D)J , Anciano , Anciano de 80 o más Años , Células Clonales , Humanos , Quimioterapia de Inducción , Mieloma Múltiple/tratamiento farmacológico , Pronóstico
9.
Am J Hematol ; 87(6): 579-87, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22495885

RESUMEN

Chromosomal abnormalities in plasma cells (PCs) from multiple myeloma (MM) provide a clonal signature to identify malignant cells. BM-lymphocytes from MM aspirates, defined by stringent criteria, were screened for the same chromosomal abnormalities as autologous PCs, including translocations, deletions, and amplifications. For 200 MM patients, we evaluated BM mononuclear cells to identify lymphocytes and autologous PCs on the same slide, followed by interphase fluorescence in situ hybridization to characterize their chromosomal abnormalities. Of all patients having a given chromosomal abnormality(s) in PCs, 45% showed that same abnormality(s) in 2-37% (median = 5%) of BM-lymphocytes. Most translocations, amplifications, and deletions found in MM PCs were also detected in lymphocytes, above the healthy-donor "cut-off." In patients having chromosomally abnormal CD20(-) PCs, chromosomally abnormal lymphocytes were found among CD20+ cells confirming them as B cells. Exceptions were amplification of 1q21 or p53 deletion, which characterize PCs but were undetectable in BM-lymphocytes, suggesting that processes leading to these abnormalities may be exclusive to PCs. For a set of 75 patients whose BM-lymphocytes and PCs were analyzed by all six probe sets, 58% of those with abnormal PC also had abnormal BM-lymphocytes harboring from one to five different abnormalities. Confirming the clinical significance of chromosomally abnormal BM-lymphocytes, MM patients having abnormalities in both lymphocytes and PC had significantly worse survival than those with abnormalities only in PC (HR = 2.68). The presence of at least one chromosomal abnormality in BM-lymphocytes appears to have greater clinical significance than particular abnormalities. Chromosomally abnormal BM-lymphocytes correlate with poor outcome and by extrapolation with more aggressive disease.


Asunto(s)
Células de la Médula Ósea/ultraestructura , Aberraciones Cromosómicas , Linfocitos/ultraestructura , Mieloma Múltiple/ultraestructura , Células Plasmáticas/ultraestructura , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD20/análisis , Deleción Cromosómica , Células Clonales/ultraestructura , Femenino , Humanos , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/mortalidad , Células Madre Neoplásicas/ultraestructura , Modelos de Riesgos Proporcionales , Muestreo , Translocación Genética , Trisomía
10.
J Cell Biochem ; 109(6): 1085-94, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20127714

RESUMEN

Multiple myeloma (MM) is characterized by karyotypic instability, including chromosomal translocations involving the IGH locus. MM cells display a promiscuity of translocation partners, only some of which are recurrent. We propose that several factors, including temporal and spatial nuclear positioning of potential partner loci, "off-target" IGH diversification mechanisms, and aberrant repair pathways contribute to the promiscuity of translocation partners in MM. We speculate that in MM, IGH diversification processes [V(D)J recombination, somatic hypermutation, and class switch recombination] in B cells may not be restricted to specific stages of B-cell development or within specific immune tissues, but may occur in different temporal "windows." Before or during MM evolution, off-target activities of the enzymes involved in IGH modification processes may contribute to the generation of double-strand breaks (DSB) in translocation partner loci. In the parent B cells from which MM originates, spatial proximity within the nucleus of IGH and potential translocation partners contributes to the selection of a translocation partner and the clinical frequency at which a specific translocation occurs. The spatial proximity of IGH and specific translocation partners may be temporal and contribute not only to partner selection but also to the promiscuity of partners seen in MM. Lastly, aberrant repair mechanisms in MM progenitors (including the possibility that a Ku 86 variant allows for positional instability at DSBs) may also contribute to the promiscuity of chromosome translocation partners in MM.


Asunto(s)
Mieloma Múltiple/genética , Translocación Genética/genética , Animales , Posicionamiento de Cromosoma/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Modelos Biológicos
11.
Blood ; 112(7): 2935-45, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18535198

RESUMEN

Although the in vitro expansion of the multiple myeloma (MM) clone has been unsuccessful, in a novel three-dimensional (3-D) culture model of reconstructed bone marrow (BM, n = 48) and mobilized blood autografts (n = 14) presented here, the entire MM clone proliferates and undergoes up to 17-fold expansion of malignant cells harboring the clonotypic IgH VDJ and characteristic chromosomal rearrangements. In this system, MM clone expands in a reconstructed microenvironment that is ideally suited for testing specificity of anti-MM therapeutics. In the 3-D model, melphalan and bortezomib had distinct targets, with melphalan targeting the hematopoietic, but not stromal com-partment. Bortezomib targeted only CD138(+)CD56(+) MM plasma cells. The localization of nonproliferating cells to the reconstructed endosteum, in contact with N-cadherin-positive stroma, suggested the presence of MM-cancer stem cells. These drug-resistant CD20(+) cells were enriched more than 10-fold by melphalan treatment, exhibited self-renewal, and generated clonotypic B and plasma cell progeny in colony forming unit assays. This is the first molecularly verified demonstration of proliferation in vitro by ex vivo MM cells. The 3-D culture provides a novel biologically relevant preclinical model for evaluating therapeutic vulnerabilities of all compartments of the MM clone, including presumptive drug-resistant MM stem cells.


Asunto(s)
Modelos Biológicos , Mieloma Múltiple/terapia , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Ácidos Borónicos/farmacología , Bortezomib , Proliferación Celular/efectos de los fármacos , Aberraciones Cromosómicas , Células Clonales , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Humanos , Melfalán/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/patología , Pirazinas/farmacología , Ratas , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Células Tumorales Cultivadas
12.
Blood ; 112(5): 1894-903, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18577707

RESUMEN

Multiple myeloma (MM) is a cancer of plasma cells (PCs) expressing immunoglobulin heavy chain (IgH) postswitch isotypes. The discovery of earlier stage cells related to postswitch PCs, called preswitch clonotypic IgM (cIgM) cells led to the hypothesis that cIgM cells may be MM progenitors, replenishing the tumor throughout malignancy. cIgM cells may do this by undergoing class switch recombination (CSR), a process detectable in postswitch PCs as multiple IgH switch junctions associated with a single clonotypic IgH V/D/J. We addressed this with a specific clonotypic-switch polymerase chain reaction (PCR), informative for 32 of 41 cases. Here we made 2 significant discoveries: (1) in all cases, we detected only a single clonotypic switch fragment that persists over time (1-7.6 years), and (2) we detected ongoing mutation upstream of the switch junction in 5 of 6 patients, often targeting the intronic enhancer, a key control region in IgH expression. The presence of a single, unchanging clonotypic switch junction suggests that cIgM cells are not MM-PC progenitors; rather, postswitch PCs arise from a single cIgM cell, and MM-PC progenitors reside in the postswitch population. Furthermore, mutations revealed here provide a new marker to identify MM-PC progenitors and aggressive clones that evolve throughout malignancy.


Asunto(s)
Cambio de Clase de Inmunoglobulina , Región de Cambio de la Inmunoglobulina , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mutación , Secuencia de Bases , Cartilla de ADN/genética , ADN de Neoplasias/genética , Humanos , Datos de Secuencia Molecular , Células Madre Neoplásicas/inmunología , Células Plasmáticas/inmunología , Reacción en Cadena de la Polimerasa , Homología de Secuencia de Ácido Nucleico , Hipermutación Somática de Inmunoglobulina , Factores de Tiempo
13.
Blood ; 112(13): 5111-21, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18815290

RESUMEN

To characterize genetic contributions toward aberrant splicing of the hyaluronan synthase 1 (HAS1) gene in multiple myeloma (MM) and Waldenstrom macroglobulinemia (WM), we sequenced 3616 bp in HAS1 exons and introns involved in aberrant splicing, from 17 patients. We identified a total of 197 HAS1 genetic variations (GVs), a range of 3 to 24 GVs/patient, including 87 somatic GVs acquired in splicing regions of HAS1. Nearly all newly identified inherited and somatic GVs in MM and/or WM were absent from B chronic lymphocytic leukemia, nonmalignant disease, and healthy donors. Somatic HAS1 GVs recurred in all hematopoietic cells tested, including normal CD34(+) hematopoietic progenitor cells and T cells, or as tumor-specific GVs restricted to malignant B and plasma cells. An in vitro splicing assay confirmed that HAS1 GVs direct aberrant HAS1 intronic splicing. Recurrent somatic GVs may be enriched by strong mutational selection leading to MM and/or WM.


Asunto(s)
Glucuronosiltransferasa/genética , Mieloma Múltiple/genética , Macroglobulinemia de Waldenström/genética , Secuencia de Bases , Progresión de la Enfermedad , Exones , Variación Genética , Sistema Hematopoyético/citología , Sistema Hematopoyético/patología , Humanos , Hialuronano Sintasas , Intrones , Mieloma Múltiple/patología , Empalme del ARN/genética , Macroglobulinemia de Waldenström/patología
14.
Br J Haematol ; 144(2): 169-75, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19036114

RESUMEN

Phase 2 trials have demonstrated that bortezomib +/- dexamethasone is safe and effective in relapsed multiple myeloma (MM). In this multicentre, open-label, phase 3b trial, 638 patients with relapsed or refractory MM (median 3 prior therapies) received bortezomib 1.3 mg/m2 on days 1, 4, 8, and 11 of a maximum of eight 3-week cycles (median 5 cycles). Dexamethasone 20 mg/d was added the day of and day after each bortezomib dose for progressive disease after > or =2 cycles or for stable disease after > or =4 cycles. Responses were assessed based on M-protein changes. Overall response rate was 67%, including 11% complete (100% M-protein reduction), 22% very good partial (75-99% reduction), 18% partial (50-74% reduction), and 16% minimal response (25-49% reduction). Dexamethasone was added in 208 patients (33%), of whom 70 (34%) showed improved response. Median time to best response of minimal response or better was 84 d. Most common grade 3/4 adverse events were thrombocytopenia (39%), neutropenia (16%), anaemia (12%), diarrhoea (7%), and peripheral neuropathy (6%). Neuropathy (any grade) was seen in 25% of the patients and led to discontinuation in 5%. Bortezomib, alone and combined with dexamethasone, is safe and effective in heavily pretreated patients with relapsed or refractory MM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Ácidos Borónicos/administración & dosificación , Dexametasona/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/sangre , Ácidos Borónicos/uso terapéutico , Bortezomib , Progresión de la Enfermedad , Esquema de Medicación , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mieloma Múltiple/sangre , Proteínas de Mieloma/análisis , Pirazinas/uso terapéutico , Recurrencia , Resultado del Tratamiento
15.
J Histochem Cytochem ; 57(3): 239-47, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19001640

RESUMEN

Multiple myeloma (MM) is an incurable B-cell malignancy that arises in the bone marrow (BM). The malignant cells within the BM have extensive interaction with the structural components of their microenvironment. It has been previously shown that the interactions between MM cells and the BM extracellular matrix (ECM) proteins contribute to drug resistance. To understand the underlying causes of adhesion-mediated drug resistance in MM, the components of human BM ECM available for interactions with MM cells must be characterized. We analyzed the expression and localization of fibronectin, laminin, and collagens I and IV in the core biopsies of normal donors and patients with monoclonal gammopathy of undetermined significance (MGUS) or MM. In addition, we compared the patterns of ECM expression in MM patients with low-, mid-, and high-level plasmacytosis of the BM. Although expression of laminin was the same for all groups tested, levels of fibronectin and collagen I were reduced in MM patients with high-level plasmacytosis. Expression of collagen IV in the BM of MGUS and MM patients was higher than in the BM from normal donors. Compared with the plasma cells isolated from the patients with low- and mid-level plasmacytosis, sorted CD138(+) plasma cells from MM patients with high-level plasmacytosis overexpressed collagen IV. Our findings show that, compared with normal controls, the ECM composition of the bone, endosteum, and BM is aberrant in patients with MM, further establishing ECM as a key player in the MM disease process.


Asunto(s)
Colágeno Tipo IV/biosíntesis , Colágeno Tipo I/biosíntesis , Fibronectinas/biosíntesis , Mieloma Múltiple/metabolismo , Paraproteinemias/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Médula Ósea/metabolismo , Huesos/metabolismo , Matriz Extracelular/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Plasmáticas/metabolismo
16.
Clin Lymphoma Myeloma ; 9(1): 30-2, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19362966

RESUMEN

The genetic factors that lead to WM are mostly unknown but are likely to involve inherited polymorphisms that might be markers of increased risk for developing WM, and somatic mutations that might be acquired during the events leading to oncogenesis and cancer progression. By intensive sequencing of the hyaluronan synthase 1 (HAS1) gene in malignant and normal cells from patients with WM, we have identified both types of mutation in HAS1 exons and introns. Acquired HAS1 mutations are found in malignant cells as well as presumptively nonmalignant CD34+ progenitor cells. This suggests that acquired HAS1 mutations precede frank malignancy and might contribute to the initial transforming events in WM as well as to disease progression.


Asunto(s)
Glucuronosiltransferasa/genética , Macroglobulinemia de Waldenström/genética , Progresión de la Enfermedad , Variación Genética , Humanos , Hialuronano Sintasas , Macroglobulinemia de Waldenström/enzimología , Macroglobulinemia de Waldenström/patología
17.
Br J Haematol ; 140(3): 295-302, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18076711

RESUMEN

Aurora kinases are potential targets for cancer therapy. Previous studies have validated Aurora kinase A as a therapeutic target in multiple myeloma (MM), and have demonstrated in vitro anti-myeloma effects of small molecule Aurora kinase inhibitors that inhibit both Aurora A and B. This study demonstrated that Aurora B kinase was strongly expressed in myeloma cell lines and primary plasma cells. The selective Aurora B inhibitor AZD1152-induced apoptotic death in myeloma cell lines at nanomolar concentrations, with a cell cycle phenotype consistent with that reported previously for Aurora B inhibition. In some cases, AZD1152 in combination with dexamethasone showed increased anti-myeloma activity compared with the use of either agent alone. AZD1152 was active against sorted CD138(+) BM plasma cells from myeloma patients but also, as expected, was toxic to CD138(-) marrow cells from the same patients. In a murine myeloma xenograft model, AZD1152-inhibited tumour growth at well-tolerated doses and induced cell death in established tumours, with associated mild, transient leucopenia. AZD1152 shows promise in these preclinical studies as a novel treatment for MM.


Asunto(s)
Antineoplásicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Organofosfatos/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/uso terapéutico , Animales , Apoptosis , Aurora Quinasa A , Aurora Quinasa B , Aurora Quinasas , Línea Celular Tumoral , Citometría de Flujo , Humanos , Inmunofenotipificación , Ratones , Ratones SCID , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Células Plasmáticas/enzimología , Células Plasmáticas/inmunología , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sindecano-1/análisis , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Clin Cancer Res ; 13(7): 2005-13, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17404080

RESUMEN

PURPOSE: Malignant B lineage cells in Waldenstrom's macroglobulinemia (WM) express a unique clonotypic IgM VDJ. The occurrence of biclonal B cells and their clonal relationships were characterized. EXPERIMENTAL DESIGN: Bone marrow and blood from 20 WM patients were analyzed for clonotypic VDJ sequences, clonal B-cell frequencies, and the complementary determining region 3 profile. RESULTS: Two different clonotypic VDJ sequences were identified in 4 of 20 WM. In two cases, partner clones had different VDJ rearrangements, with one clonotypic signature in bone marrow and a second in blood. For both cases, the bone marrow clone was hypermutated, whereas the blood clone was germ line or minimally mutated. In two other cases, partner clones shared a common VDJ rearrangement but had different patterns of somatic mutations. They lacked intraclonal diversity and were more abundant in bone marrow than in blood. VDJ mutation profiles suggested they arose from a common IgM progenitor. Single-cell analysis in one case indicated the partner clones were reciprocally expressed, following rules of allelic exclusion. CONCLUSIONS: The existence of two B-cell clones having distinct VDJ sequences is common in WM, suggesting that frequent transformation events may occur. In two cases, the partner clones had distinct tissue distributions in either blood or bone marrow, were of different immunoglobulin isotypes, and in one case exhibited differential response to therapy. The contributions of each clone are unknown. Their presence suggests that WM may involve a background of molecular and cellular events leading to emergence of one or more malignant clones.


Asunto(s)
Linfocitos B/inmunología , Regiones Determinantes de Complementariedad/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Inmunoglobulina M/genética , Macroglobulinemia de Waldenström/genética , Anciano , Linfocitos B/citología , Secuencia de Bases , Linaje de la Célula , Separación Celular , Células Clonales , Electroforesis Capilar , Femenino , Citometría de Flujo , Reordenamiento Génico de Cadena Pesada de Linfocito B , Células Madre Hematopoyéticas , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Mol Diagn ; 9(3): 358-67, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17591936

RESUMEN

Diagnosis platforms incorporating low-cost microfluidic chips enable sensitive, rapid, and accurate genetic analysis that could facilitate customized therapies tailored to match the vulnerabilities of any types of cancer. Using ex vivo cancer cells, we have detected the unique molecular signature and a chromosomal translocation in multiple myeloma. Multiple myeloma is characterized by IgH rearrangements and translocations that enable unequivocal identification of malignant cells, detected here with integrated microfluidic chips incorporating genetic amplification via reverse transcriptase-polymerase chain reaction and capillary electrophoresis. On microfluidic chips, we demonstrated accurate and versatile detection of molecular signatures in individual cancer cells, with value for monitoring response to therapy, detecting residual cancer cells that mediate relapse, and evaluating prognosis. Thus, testing for two clinically important molecular biomarkers, the IgH VDJ signature and hybrid transcripts signaling the t(4;14) chro-mosomal translocation, with predictive value in diagnosis, treatment decisions, and monitoring has been efficiently implemented on a miniaturized microfluidic system.


Asunto(s)
Cromosomas Humanos Par 14 , Cromosomas Humanos Par 4 , Técnicas Analíticas Microfluídicas/métodos , Mieloma Múltiple/diagnóstico , Proteínas de Fusión Oncogénica/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Translocación Genética , Algoritmos , Médula Ósea/metabolismo , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Genes de las Cadenas Pesadas de las Inmunoglobulinas , Genes Relacionados con las Neoplasias , Humanos , Monitoreo Fisiológico/métodos , Mieloma Múltiple/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Sensibilidad y Especificidad
20.
Cancer Res ; 65(3): 850-60, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15705883

RESUMEN

Elevated expression of receptor for hyaluronan-mediated motility (RHAMM) within ex vivo diagnostic multiple myeloma plasma cells predicts for aggressive disease and patient survival. Here, we investigate the relationship between RHAMM and centrosomal abnormalities within multiple myeloma patient samples. We report that myeloma patient samples contain pervasive structural and numerical centrosomal abnormalities. Structural, but not numerical, centrosomal abnormalities strongly correlate with elevated RHAMM expression. As others have shown that excess pericentriolar material strongly associates with abnormal mitoses, we modeled centrosomal abnormalities with exogenous RHAMM overexpression. RHAMM overexpression in vitro resulted in centrosomal and mitotic defects. To elucidate a mechanism for RHAMM-mediated spindle defects, we further investigated RHAMM mitotic function. RHAMM mitotic localization mirrors that of targeting protein for Xklp2 (TPX2), and RHAMM interacts with the spindle assembly factors dynein and TPX2. Like TPX2, RHAMM expression is up-regulated during mitosis. Moreover, inhibition of function experiments reveals that RHAMM and TPX2 functions converge to maintain spindle integrity after spindle assembly. We postulate that augmentation of RHAMM expression within human cancers, including myeloma, can directly affect centrosomal structure and spindle integrity and potentially modulate apoptotic and cell cycle progression pathways.


Asunto(s)
Movimiento Celular/fisiología , Centrosoma/fisiología , Aberraciones Cromosómicas , Proteínas de la Matriz Extracelular/fisiología , Receptores de Hialuranos/fisiología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Huso Acromático/fisiología , Ciclo Celular/genética , Ciclo Celular/fisiología , Movimiento Celular/genética , Centrosoma/metabolismo , Centrosoma/patología , Segregación Cromosómica/fisiología , Dineínas/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Células HeLa , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Mitosis/genética , Mieloma Múltiple/metabolismo , Huso Acromático/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA