Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Genes Immun ; 15(1): 54-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24153350

RESUMEN

Sjogren's syndrome (SS) is a chronic autoimmune disorder with the highest risk for lymphoma development among all autoimmune diseases. In order to evaluate whether the presence of the recently described MYD88 L265P mutation in patients with Waldenström's macroglobulinemia (WM) is contributory to SS-associated lymphomagenesis, a quantitative allele-specific PCR method was performed in peripheral blood derived from 90 SS patients as well as in minor salivary gland tissues derived from 12 primary SS patients with or without lymphoma. MYD88 L265P was not detected in either of the samples tested. Although the absence of the MyD88 L265P somatic mutation in our SS cohort does not exclude a common germline susceptibility gene in SS, it might suggest a distinct operating pathogenetic mechanism in SS-related lymphoma compared with WM and other hematological malignancies.


Asunto(s)
Linfoma/genética , Mutación , Factor 88 de Diferenciación Mieloide/genética , Síndrome de Sjögren/genética , Humanos , Linfoma/etiología , Reacción en Cadena de la Polimerasa , Glándulas Salivales/fisiología , Síndrome de Sjögren/complicaciones , Síndrome de Sjögren/patología
2.
Semin Hematol ; 60(2): 107-112, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37099029

RESUMEN

Consensus Panel 5 (CP5) of the 11th International Workshop on Waldenstrom's Macroglobulinemia (IWWM-11; held in October 2022) was tasked with reviewing the current data on the coronavirus disease-2019 (COVID-19) prophylaxis and management in patients with Waldenstrom's Macroglobulinemia (WM). The key recommendations from IWWM-11 CP5 included the following: Booster vaccines for SARS-CoV-2 should be recommended to all patients with WM. Variant-specific booster vaccines, such as the bivalent vaccine for the ancestral Wuhan strain and the Omicron BA.4.5 strain, are important as novel mutants emerge and become dominant in the community. A temporary interruption in Bruton's Tyrosine Kinase-inhibitor (BTKi) or chemoimmunotherapy before vaccination might be considered. Patients under treatment with rituximab or BTK-inhibitors have lower antibody responses against SARS-CoV-2; thus, they should continue to follow preventive measures, including mask wearing and avoiding crowded places. Patients with WM are candidates for preexposure prophylaxis, if available and relevant to the dominant SARS-CoV-2 strains in a specific area. Oral antivirals should be offered to all symptomatic WM patients with mild to moderate COVID-19 regardless of vaccination, disease status or treatment, as soon as possible after the positive test and within 5 days of COVID-19-related symptom onset. Coadministration of ibrutinib or venetoclax with ritonavir should be avoided. In these patients, remdesivir offers an effective alternative. Patients with asymptomatic or oligosymptomatic COVID-19 should not interrupt treatment with a BTK inhibitor. Infection prophylaxis is essential in patients with WM and include general preventive measures, prophylaxis with antivirals and vaccination against common pathogens including SARS-CoV-2, influenza, and S. pneumoniae.


Asunto(s)
COVID-19 , Macroglobulinemia de Waldenström , Humanos , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/prevención & control , Macroglobulinemia de Waldenström/diagnóstico , Vacunas contra la COVID-19 , Consenso , SARS-CoV-2 , Antivirales/uso terapéutico
3.
Semin Hematol ; 60(2): 90-96, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37099028

RESUMEN

Apart from the MYD88L265P mutation, extensive information exists on the molecular mechanisms in Waldenström's Macroglobulinemia and its potential utility in the diagnosis and treatment tailoring. However, no consensus recommendations are yet available. Consensus Panel 3 (CP3) of the 11th International Workshop on Waldenström's Macroglobulinemia (IWWM-11) was tasked with reviewing the current molecular necessities and best way to access the minimum data required for a correct diagnosis and monitoring. Key recommendations from IWWM-11 CP3 included: (1) molecular studies are warranted for patients in whom therapy is going to be started; such studies should also be done in those whose bone marrow (BM) material is sampled based on clinical issues; (2) molecular studies considered essential for these situations are those that clarify the status of 6q and 17p chromosomes, and MYD88, CXCR4, and TP53 genes. These tests in other situations, and/or other tests, are considered optional; (3) independently of the use of more sensitive and/or specific techniques, the minimum requirements are allele specific polymerase chain reaction for MYD88L265P and CXCR4S338X using whole BM, and fluorescence in situ hybridization for 6q and 17p and sequencing for CXCR4 and TP53 using CD19+ enriched BM; (4) these requirements refer to all patients; therefore, sample should be sent to specialized centers.


Asunto(s)
Macroglobulinemia de Waldenström , Humanos , Macroglobulinemia de Waldenström/diagnóstico , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/terapia , Factor 88 de Diferenciación Mieloide/genética , Hibridación Fluorescente in Situ , Mutación
4.
Semin Hematol ; 60(2): 118-124, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37099031

RESUMEN

Recent advances in the understanding of Waldenström macroglobulinemia (WM) biology have impacted the development of effective novel agents and improved our knowledge of how the genomic background of WM may influence selection of therapy. Consensus Panel 7 (CP7) of the 11th International Workshop on WM was convened to examine the current generation of completed and ongoing clinical trials involving novel agents, consider updated data on WM genomics, and make recommendations on the design and prioritization of future clinical trials. CP7 considers limited duration and novel-novel agent combinations to be the priority for the next generation of clinical trials. Evaluation of MYD88, CXCR4 and TP53 at baseline in the context of clinical trials is crucial. The common chemoimmunotherapy backbones, bendamustine-rituximab (BR) and dexamethasone, rituximab and cyclophosphamide (DRC), may be considered standard-of-care for the frontline comparative studies. Key unanswered questions include the definition of frailty in WM; the importance of attaining a very good partial response or better (≥VGPR), within stipulated time frame, in determining survival outcomes; and the optimal treatment of WM populations with special needs.


Asunto(s)
Macroglobulinemia de Waldenström , Humanos , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/genética , Rituximab/uso terapéutico , Consenso , Ciclofosfamida/uso terapéutico , Clorhidrato de Bendamustina/uso terapéutico
5.
Semin Hematol ; 60(2): 80-89, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37147252

RESUMEN

The consensus panel 2 (CP2) of the 11th International Workshop on Waldenström's macroglobulinemia (IWWM-11) has reviewed and incorporated current data to update the recommendations for treatment approaches in patients with relapsed or refractory WM (RRWM). The key recommendations from IWWM-11 CP2 include: (1) Chemoimmunotherapy (CIT) and/or a covalent Bruton tyrosine kinase (cBTKi) strategies are important options; their use should reflect the prior upfront strategy and are subject to their availability. (2) In selecting treatment, biological age, co-morbidities and fitness are important; nature of relapse, disease phenotype and WM-related complications, patient preferences and hematopoietic reserve are also critical factors while the composition of the BM disease and mutational status (MYD88, CXCR4, TP53) should also be noted. (3) The trigger for initiating treatment in RRWM should utilize knowledge of patients' prior disease characteristics to avoid unnecessary delays. (4) Risk factors for cBTKi related toxicities (cardiovascular dysfunction, bleeding risk and concurrent medication) should be addressed when choosing cBTKi. Mutational status (MYD88, CXCR4) may influence the cBTKi efficacy, and the role of TP53 disruptions requires further study) in the event of cBTKi failure dose intensity could be up titrated subject to toxicities. Options after BTKi failure include CIT with a non-cross-reactive regimen to one previously used CIT, addition of anti-CD20 antibody to BTKi, switching to a newer cBTKi or non-covalent BTKi, proteasome inhibitors, BCL-2 inhibitors, and new anti-CD20 combinations are additional options. Clinical trial participation should be encouraged for all patients with RRWM.


Asunto(s)
Antineoplásicos , Macroglobulinemia de Waldenström , Humanos , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/genética , Factor 88 de Diferenciación Mieloide/genética , Consenso , Recurrencia Local de Neoplasia/inducido químicamente , Recurrencia Local de Neoplasia/tratamiento farmacológico , Antineoplásicos/uso terapéutico
6.
J Clin Invest ; 101(6): 1379-88, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9502780

RESUMEN

Previous studies have shown that triggering multiple myeloma (MM) cells via CD40 induces IL-6-mediated autocrine growth as well as increased expression of cell surface adhesion molecules including CD11a, CD11b, CD11c, and CD18. In this study, we generated the 5E2 mAb which targets an antigen that is induced upon CD40 ligand (CD40L) activation of MM cells. Immunofluorescence, immunoprecipitation, and protein sequencing studies identified the target antigen of 5E2 mAb as the 86-kD subunit of the Ku autoantigen. We demonstrate that increased cell surface expression of Ku on CD40L-treated cells is due to migration of Ku from the cytoplasm to the cell surface membrane. Moreover, cell surface Ku on CD40L-treated MM cells mediates homotypic adhesion of tumor cells, as well as heterotypic adhesion of tumor cells to bone marrow stromal cells and to human fibronectin; and 5E2 mAb abrogates IL-6 secretion triggered by tumor cell adherence to bone marrow stromal cells. These data suggest that CD40L treatment induces a shift of Ku from the cytoplasm to the cell surface, and are the first to show that Ku functions as an adhesion molecule. They further suggest that cell surface Ku may play a role in both autocrine and paracrine IL-6-mediated MM cell growth and survival.


Asunto(s)
Antígenos Nucleares , Autoanticuerpos/inmunología , ADN Helicasas , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Western Blotting , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Antígenos CD40/inmunología , Ligando de CD40 , Adhesión Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citoplasma/inmunología , Citoplasma/metabolismo , Proteínas de Unión al ADN/aislamiento & purificación , Fibronectinas/metabolismo , Citometría de Flujo , Humanos , Interleucina-6/metabolismo , Autoantígeno Ku , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas Nucleares/aislamiento & purificación , Pruebas de Precipitina , Células del Estroma/inmunología , Células del Estroma/metabolismo , Transfección , Células Tumorales Cultivadas
7.
Leukemia ; 30(5): 1116-25, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26867669

RESUMEN

Waldenström's macroglobulinemia (WM) is a B-cell non-Hodgkin's lymphoma (B-NHL) characterized by immunoglobulin M (IgM) monoclonal gammopathy and the medullary expansion of clonal lymphoplasmacytic cells. Neoplastic transformation has been partially attributed to hyperactive MYD88 signaling, secondary to the MYD88 L265P mutation, occurring in the majority of WM patients. Nevertheless, the presence of chronic active B-cell receptor (BCR) signaling, a feature of multiple IgM+ B-NHL, remains a subject of speculation in WM. Here, we interrogated the BCR signaling capacity of primary WM cells by utilizing multiparametric phosphoflow cytometry and found heightened basal phosphorylation of BCR-related signaling proteins, and augmented phosphoresponses on surface IgM (sIgM) crosslinking, compared with normal B cells. In support of those findings we observed high sIgM expression and loss of phosphatase activity in WM cells, which could both lead to signaling potentiation in clonal cells. Finally, led by the high-signaling heterogeneity among WM samples, we generated patient-specific phosphosignatures, which subclassified patients into a 'high' and a 'healthy-like' signaling group, with the second corresponding to patients with a more indolent clinical phenotype. These findings support the presence of chronic active BCR signaling in WM while providing a link between differential BCR signaling utilization and distinct clinical WM subgroups.


Asunto(s)
Linfocitos B/patología , Receptores de Antígenos de Linfocitos B/fisiología , Transducción de Señal , Macroglobulinemia de Waldenström/patología , Células Clonales/patología , Femenino , Humanos , Inmunoglobulina M/metabolismo , Masculino , Fosforilación
8.
Blood Cancer J ; 6: e380, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26771806

RESUMEN

Deregulated microRNA (miR)/transcription factor (TF)-based networks represent a hallmark of cancer. We report here a novel c-Myc/miR-23b/Sp1 feed-forward loop with a critical role in multiple myeloma (MM) and Waldenstrom's macroglobulinemia (WM) cell growth and survival. We have found miR-23b to be downregulated in MM and WM cells especially in the presence of components of the tumor bone marrow milieu. Promoter methylation is one mechanism of miR-23b suppression in myeloma. In gain-of-function studies using miR-23b mimics-transfected or in miR-23b-stably expressing MM and WM cell lines, we observed a significant decrease in cell proliferation and survival, along with induction of caspase-3/7 activity over time, thus supporting a tumor suppressor role for miR-23b. At the molecular level, miR-23b targeted Sp1 3'UTR and significantly reduced Sp1-driven nuclear factor-κB activity. Finally, c-Myc, an important oncogenic transcription factor known to stimulate MM cell proliferation, transcriptionally repressed miR-23b. Thus MYC-dependent miR-23b repression in myeloma cells may promote activation of oncogenic Sp1-mediated signaling, representing the first feed-forward loop with critical growth and survival role in myeloma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , MicroARNs/genética , Mieloma Múltiple/genética , Proteínas Proto-Oncogénicas c-myc/genética , Factor de Transcripción Sp1/genética , Animales , Secuencia de Bases , Sitios de Unión , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Metilación de ADN , Modelos Animales de Enfermedad , Regulación hacia Abajo , Expresión Génica , Perfilación de la Expresión Génica , Silenciador del Gen , Genes Reporteros , Humanos , MicroARNs/química , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/química , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/genética , Factor de Transcripción Sp1/química , Factor de Transcripción Sp1/metabolismo
9.
Leukemia ; 6(10): 1036-42, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1405756

RESUMEN

Post-transfusional hepatitis is often a complication in patients with acute myelogenous leukemia (AML) in whom survival is paradoxically prolonged. The etiology is unknown. In previous studies, we showed that impaired hepatic endotoxin (lipopolysaccharide, LPS) clearance in patients with acute viral hepatitis A, B, or C versus controls results in endotoxemia and tumor necrosis factor alpha (TNF-alpha) release. TNF-alpha mediates anti-proliferative and differentiating effects in AML cell lines. Interferon-gamma (IFN-gamma) released in acute viral hepatitis, acts in synergy with TNF-alpha. HL60, KG1, and U937 AML cells treated 3, 6, and 9 days with physiologically attainable TNF-alpha (10 U/ml), IFN-gamma (100 U/ml) and LPS (10 ng/ml) levels, have significantly diminished viability and cell growth versus controls. Treatment of HL60 AML cells with LPS/TNF-alpha/IFN-gamma also resulted in significantly increased monocytic pathway differentiation not seen with KG1 or U937 AML cells. HL60 AML cells treated with TNF-alpha/IFN-gamma for 6 days released endogenous TNF-alpha (1.57 U/10(6) cells) upon LPS stimulation compared to less than 0.01 U/10(6) cells in non-LPS-stimulated TNF-alpha/IFN-gamma-treated cells or untreated cells (p less than 0.0001). Untreated HL60 AML cells co-cultured with HL60 cells pretreated for 6 days with TNF-alpha/IFN-gamma and then subjected to LPS stimulation had significantly diminished cell growth compared to controls (p less than 0.0001). This effect could be reversed with anti-TNF-alpha antibody, supporting the concept that endogenous TNF-alpha release by LPS/TNF-alpha/IFN-gamma treated HL60 AML cells may act by paracrine means to suppress growth of other AML cells. The beneficial effects of post-transfusional hepatitis in AML patients may be mediated via LPS/TNF-alpha/IFN-gamma-induced AML cell growth suppression and/or terminal differentiation in which AML cells participate by releasing TNF-alpha after being acted upon by LPS/TNF-alpha/IFN-gamma. Endogenously released TNF-alpha might then act by autocrine/paracrine means to mediate further suppression and terminal differentiation.


Asunto(s)
División Celular/efectos de los fármacos , Hepatitis/complicaciones , Interferón gamma/administración & dosificación , Leucemia Mieloide Aguda/patología , Lipopolisacáridos/administración & dosificación , Factor de Necrosis Tumoral alfa/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Hepatitis/fisiopatología , Humanos , Técnicas In Vitro , Leucemia Mieloide Aguda/complicaciones , Leucemia Mieloide Aguda/fisiopatología , Leucemia Mieloide Aguda/terapia , Proteínas Recombinantes , Reacción a la Transfusión , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
10.
Leukemia ; 6 Suppl 3: 141S-145S, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1602811

RESUMEN

The microtubule (MT) network of the cytoskeleton has been implicated as a mediator of cellular signal transduction; disorganization of this network may allow for mitogenesis. In previous work, loss of MT network organization in human MOLT4 and HUT78 T-cell leukemias was demonstrated in contrast to an organized "spoke-wheel-like arrangement" in normal human T-lymphocytes. In this study, loss of MT network organization was shown in several representative acute myeloid leukemia (AML) cell lines: KG1 myeloblastic, HL60 promyelocytic, and U937 myelomonocytic cells. Re-organization of the MT network was observed in HL60 and U937 AML cells treated with combined lipopolysaccharide (LPS), tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma). This re-organization paralleled earlier work which showed this combination was effective in inducing monocytic pathway differentiation and growth restraint in HL60 cells, and growth restraint in U937 cells. In contrast, KG1 cells exhibited growth restraint, but did not re-organize with LPS/TNF-alpha/IFN-gamma treatment. These results are consistent with a role for the MT network in mitogenesis. Loss of MT network organization appeared to parallel the neoplastic phenotype in three AML cell lines, whereas MT network re-organization accompanied recovery of growth control in 2 of 3 AML cell lines.


Asunto(s)
Interferón gamma/uso terapéutico , Leucemia Mieloide Aguda/patología , Leucemia Mielomonocítica Aguda/patología , Leucemia Promielocítica Aguda/patología , Lipopolisacáridos/uso terapéutico , Microtúbulos/ultraestructura , Factor de Necrosis Tumoral alfa/uso terapéutico , Leucemia Mieloide Aguda/terapia , Leucemia Mielomonocítica Aguda/terapia , Leucemia Promielocítica Aguda/terapia , Microtúbulos/efectos de los fármacos , Células Tumorales Cultivadas
11.
Leukemia ; 11(11): 1957-63, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9369432

RESUMEN

Interleukin-6 (IL-6) promotes growth of human multiple myeloma (MM) cells via phosphorylation of retinoblastoma protein (pRB). We therefore examined the kinetics of cyclin-dependent kinase 4 (CDK4), p16INK4A, and pRB activation during IL-6-mediated patient MM cell growth compared with growth of IL-6 unresponsive patient plasma cell leukemia (PCL) cells. CDK4 protein was more strongly expressed in PCL cells than in MM cells. On the other hand, p16 protein was present in MM cells but undetectable in PCL cells. Interestingly, IL-6 induced peak proliferation of MM cells at days 1-3, with a return to baseline levels of DNA synthesis by days 6-9 in spite of replenishing IL-6. In these cells, IL-6 triggered a sustained increase in CDK4 by day 1 and a gradual increase in p16 to day 9. The progressive increase in p16 without further increments in CDK4 resulted in a shift from cyclin D2-CDK4/CDK6 binding at days 1-3 to p16-CDK4/CDK6 complex formation at days 6-9. Both phosphorylated pRB and dephosphorylated pRB were present initially in patient MM cells; IL-6 triggered a shift to phosphorylated pRB and G1 to S transition at days 1-3, with return to baseline levels of dephosphorylated pRB and related G1 growth arrest by day 9. No similar changes in CDK4, p16, or cell cycle profile were observed in IL-6 nonresponsive PCL cells. Our data therefore suggest a feedback mechanism in IL-6-mediated MM cell growth which is absent in IL-6 nonresponsive PCL cells.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Interleucina-6/farmacología , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas , Proteína de Retinoblastoma/metabolismo , Recuento de Células/efectos de los fármacos , Técnicas de Cultivo de Célula , División Celular/efectos de los fármacos , Ciclina D2 , Quinasa 4 Dependiente de la Ciclina , Ciclinas/metabolismo , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Mieloma Múltiple/metabolismo , Células Tumorales Cultivadas
12.
Leukemia ; 15(12): 1950-61, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11753617

RESUMEN

Increased angiogenesis has recently been recognized in active multiple myeloma (MM). Since vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) are two key mediators of angiogenesis, we characterized the production of VEGF, b-FGF and interleukin-6 (IL-6) (a MM growth and survival factor) in MM cell lines and Epstein-Barr virus (EBV) transformed B cell lines from MM patients, patient MM cells, as well as bone marrow stromal cells (BMSCs) from normal healthy donors and MM patients. We detected secretion of VEGF, but no bFGF and IL-6, in MM cell lines (MM.1S, RPMI 8226 and U266); EBV transformed B cell lines from MM patients (IM-9, HS-Sultan and ARH77); MM cell lines resistant to doxorubicin (RPMI-DOX40), mitoxantrone (RPMI-MR20), melphalan (RPMI-LR5) and dexamethasone (MM.1R); and patient MM cells (MM1 and MM2). BMSCs from MM patients and normal donors secreted VEGF, b-FGF and IL-6. Importantly, when MM cells were adhered to BMSCs, there was a significant increase in VEGF (1.5- to 3.1-fold) and IL-6 (1.9- to 56-fold) secretion. In contrast, the bFGF decreased in co-cultures of BMSCs and MM cells. Paraformaldehyde fixation of BMSCs or MM cells prior to adhesion revealed that VEGF was produced both from BMSCs and MM cells, though it may come primarily from BMSCs in some cultures. IL-6 was produced exclusively in BMSCs, rather than MM cells. Moreover, when MM cells were placed in Transwell insert chambers to allow their juxtaposition to BMSCs without cell to cell contact, induction of VEGF and IL-6 secretion persisted, suggesting the importance of humoral factors. Addition of exogenous IL-6 (10 ng/ml) increased VEGF secretion by BMSCs. Conversely, VEGF (100 ng/ml) significantly increased IL-6 secretion by BMSCs. Moreover, anti-human VEGF (1 microg/ml) and anti-human IL-6 (10 microg/ml) neutralizing antibodies reduced IL-6 and VEGF secretion, respectively, in cultures of BMSCs alone and co-cultures of BMSCs and MM cells. Finally, thalidomide (100 microM) and its immunomodulatory analog IMiD1-CC4047 (1 microM) decreased the upregulation of IL-6 and VEGF secretion in cultures of BMSCs, MM cells and co-cultures of BMSCs with MM cells. These data demonstrate the importance of stromal-MM cell interactions in regulating VEGF and IL-6 secretion, and suggest additional mechanisms whereby thalidomide and IMiD1-CC4047 act against MM cells in the BM millieu.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Linfocinas/metabolismo , Mieloma Múltiple/patología , Células del Estroma/citología , Inhibidores de la Angiogénesis/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular , Comunicación Celular/fisiología , Técnicas de Cocultivo , Interacciones Farmacológicas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/fisiopatología , Células del Estroma/metabolismo , Células del Estroma/fisiología , Talidomida/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Clin Cancer Res ; 3(6): 1017-22, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9815779

RESUMEN

Interleukin 6 (IL-6) is a growth factor for multiple myeloma (MM) cells, yet not all MM cell lines or patient cells require IL-6 for their growth. It is well known that IL-6 activates the signal transducers and activators of transcription (stat) 1-stat3 heterodimer, stat3 homodimer, and Ras-dependent mitogen-activated protein kinase (MAPK) cascades in multiple cell systems. We have shown previously that the MAPK pathway is an important pathway for IL-6-mediated MM cell growth. In this study, we delineate the pattern of upstream MAPK cascade activation in IL-6-responsive B9 cells and in IL-6-nonresponsive U266, OCI-My5, and RPMI8226 MM cells to define sites of blockade of this pathway associated with loss of responsiveness to IL-6. In B9 cells, IL-6 triggered the following in sequence: gp130 phosphorylation, gp130-to-protein tyrosine phosphatase 1D (PTP1D) binding, PTP1D phosphorylation, PTP1D complex formation with Grb2-Son of sevenless 1 (Sos1), and Sos1 phosphorylation. gp130 phosphorylation, gp130-to-PTP1D binding, PTP1D phosphorylation, and PTP1D-to-Grb2 binding are also induced by IL-6 in all IL-6-independent MM cell lines studied. However, Grb2 is not associated with Sos1, and neither Grb2-to-Sos1 binding nor Sos1 phosphorylation is triggered by IL-6 in OCI-My5 MM cells. On the other hand, Grb2 and Sos1 are associated constitutively in U266 and RPMI8226 MM cells, but phosphorylation of Sos1 is not induced by IL-6. These data suggest that lack of Sos1 activation is associated with loss of IL-6 responsiveness in MM cell lines that grow independently of IL-6.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Interleucina-6/farmacología , Transducción de Señal/fisiología , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , División Celular , Proteína Adaptadora GRB2 , Factores de Intercambio de Guanina Nucleótido , Humanos , Interleucina-6/fisiología , Ratones , Mieloma Múltiple , Fosforilación , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas , Factores de Intercambio de Guanina Nucleótido ras
14.
Clin Cancer Res ; 6(3): 1180-9, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10741750

RESUMEN

Kaposi's sarcoma-associated herpes virus (KSHV) is associated with Kaposi's sarcoma, multicentric Castleman's disease, and body cavity-based lymphomas, settings in which human interleukin-6 (hIL-6) acts as a growth factor. The KSHV open reading frame K2 encodes for viral IL-6 (vIL-6), a protein with 25% amino acid identity to hIL-6, which can promote the growth of hIL-6-dependent cell lines. In the present study, we characterized biological sequelae and signaling cascades triggered by hIL-6 versus vIL-6 in the hIL-6-dependent MH60 and B9 cell lines. Both hIL-6 and vIL-6 induced significant increases (P < 0.01) in DNA synthesis in these cell lines in a dose-dependent fashion. Neutralizing anti-hIL-6 antibody (Ab) inhibited DNA synthesis triggered by hIL-6, without similarly affecting proliferation in response to vIL-6. On the other hand, antimouse IL-6 receptor (mIL-6R) Ab blocked response to vIL-6, but not that to hIL-6. Both hIL-6 and vIL-6 activated gp130, Janus kinase 1, signal transducers and activators of transcription-3, and mitogen-activated protein kinase in both MH60 and B9 cells. Proliferation of these cell lines in response to both hIL-6 and vIL-6 was blocked by PD98059, an inhibitor of MEK1 activation. These data suggest that MEK1 activation mediates the proliferative response to both cytokines. Finally, both hIL-6 and vIL-6 also maintained viability of serum-starved MH60 and B9 cells and blocked dexamethasone-induced apoptosis of MM.1S human myeloma cells. Further characterization of the signaling cascades mediating the growth and antiapoptotic effects of vIL-6 versus hIL-6 may help identify their unique roles in disease pathogenesis in Kaposi's sarcoma and other KSHV-associated neoplasms.


Asunto(s)
Interleucina-6/farmacología , Proteínas Proto-Oncogénicas , Transducción de Señal/efectos de los fármacos , Proteínas Virales/farmacología , Animales , Anticuerpos/farmacología , Antígenos CD/efectos de los fármacos , Antígenos CD/metabolismo , División Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Receptor gp130 de Citocinas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Dexametasona/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Herpesvirus Humano 8/genética , Humanos , Hibridomas , Interleucina-6/genética , Interleucina-6/inmunología , Janus Quinasa 1 , Janus Quinasa 2 , MAP Quinasa Quinasa 1 , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Pruebas de Neutralización , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Receptores de Interleucina-6/inmunología , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT1 , Factor de Transcripción STAT2 , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Células Tumorales Cultivadas , Tirosina/metabolismo , Proteínas Virales/genética
15.
Clin Cancer Res ; 3(11): 2173-9, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9815612

RESUMEN

Loss of p16(INK4A) (p16) expression is frequently associated with the development of epithelial and lymphoid malignancies. However, the frequency and significance of p16 abnormalities in multiple myeloma (MM) and the more aggressive phase of plasma cell leukemia (PCL) have not been well defined. Accordingly, the goal of this study was to define the expression and function of p16 in fresh samples of MM and PCL. We found that p16 protein was highly expressed in primary MM cells, although it was undetectable in fresh samples of PCL. Additionally, p16 protein was also absent in four of four MM-derived cell lines. To determine the mechanism for p16 underexpression in PCL and MM-derived cell lines, we performed PCR analysis to evaluate both gene deletion and the presence of methylation. Interestingly, the p16 gene was present and methylated in all patient PCL cells and MM cell lines, whereas it was unmethylated in patient MM cells and normal B cells. Furthermore, treatment with the demethylating agent 5-deoxyazacytidine or p16 retrofection restored p16 protein expression and induced G1 growth arrest in patient PCL cells and MM cell lines. These results suggest that inactivation of the p16 gene by methylation may be associated with decreased growth control and the development of PCL in a subset of patients with MM.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Genes p16 , Leucemia de Células Plasmáticas/genética , Mieloma Múltiple/genética , Linfocitos B/metabolismo , Linfocitos B/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Células Cultivadas , Metilación de ADN , ADN de Neoplasias/metabolismo , Humanos , Leucemia de Células Plasmáticas/metabolismo , Leucemia de Células Plasmáticas/patología , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Tumorales Cultivadas
16.
Leukemia ; 29(1): 169-76, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24912431

RESUMEN

CXCR4(WHIM) somatic mutations are common Waldenstrom's Macroglobulinemia (WM), and are associated with clinical resistance to ibrutinib. We engineered WM cells to express the most common WHIM (Warts, Hypogammaglobulinemia, Infections and Myelokathexis), CXCR(S338X) mutation in WM. Following SDF-1a stimulation, CXCR4(S338X) WM cells exhibited decreased receptor internalization, enhanced and sustained AKT kinase (AKT) and extracellular regulated kinase (ERK) signaling, decreased poly (ADP-ribose) polymerase and caspase 3 cleavage, and decreased Annexin V staining versus CXCR4 wild-type (WT) cells. CXCR4(S338X)-related signaling and survival effects were blocked by the CXCR4 inhibitor AMD3100. SDF-1a-treated CXCR4(S338X) WM cells showed sustained AKT and ERK activation and decreased apoptotic changes versus CXCR4(WT) cells following ibrutinib treatment, findings which were also reversed by AMD3100. AKT or ERK antagonists restored ibrutinib-triggered apoptotic changes in SDF-1a-treated CXCR4(S338X) WM cells demonstrating their role in SDF-1a-mediated ibrutinib resistance. Enhanced bone marrow pAKT staining was also evident in CXCR4(WHIM) versus CXCR4(WT) WM patients, and remained active despite ibrutinib therapy in CXCR4(WHIM) patients. Last, CXCR4(S338X) WM cells showed varying levels of resistance to other WM relevant therapeutics, including bendamustine, fludarabine, bortezomib and idelalisib in the presence of SDF-1a. These studies demonstrate a functional role for CXCR4(WHIM) mutations, and provide a framework for investigation of CXCR4 inhibitors in WM.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Mutación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Receptores CXCR4/genética , Macroglobulinemia de Waldenström/tratamiento farmacológico , Adenina/análogos & derivados , Activación Enzimática , Humanos , Piperidinas , Macroglobulinemia de Waldenström/genética
17.
Leukemia ; 29(12): 2338-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26139427

RESUMEN

We examined the combination of the mammalian target of rapamycin inhibitor everolimus with bortezomib and rituximab in patients with relapsed/refractory Waldenstrom macroglobulinemia (WM) in a phase I/II study. All patients received six cycles of the combination of everolimus/rituximab or everolimus/bortezomib/rituximab followed by maintenance with everolimus until progression. Forty-six patients were treated; 98% received prior rituximab and 57% received prior bortezomib. No dose-limiting toxicities were observed in the phase I. The most common treatment-related toxicities of all grades were fatigue (63%), anemia (54%), leucopenia (52%), neutropenia (48%) and diarrhea (43%). Thirty-six (78%) of the 46 patients received full dose therapy (FDT) of the three drugs. Of these 36, 2 (6%) had complete response (90% confidence interval (CI): 1-16). In all, 32/36 (89%) of patients experienced at least a minimal response (90% CI: 76-96%). The observed partial response or better response rate was 19/36 (53, 90 CI: 38-67%). For the 36 FDT patients, the median progression-free survival was 21 months (95% CI: 12-not estimable). In summary, this study demonstrates that the combination of everolimus, bortezomib and rituximab is well tolerated and achieved 89% response rate even in patients previously treated, making it a possible model of non-chemotherapeutic-based combination therapy in WM.


Asunto(s)
Macroglobulinemia de Waldenström/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Bortezomib/administración & dosificación , Bortezomib/efectos adversos , Quimioterapia Combinada , Everolimus/administración & dosificación , Everolimus/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Factor 88 de Diferenciación Mieloide/genética , Receptores CXCR4/genética , Recurrencia , Rituximab/administración & dosificación , Rituximab/efectos adversos
18.
Semin Oncol ; 27(6 Suppl 12): 79-85, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11226004

RESUMEN

CD20 is a B-cell-restricted antigen that, for the most part, is expressed from the pre-B-cell to the mature B-cell stage of B-cell differentiation. Several transcription factors regulate CD20 expression during B-cell differentiation, the most important of which appear to be PU.1 and Pip (PU.1 interacting protein). As B cells differentiate to plasma cells, CD20 expression is down-regulated, which coincides with PU.1 downregulation in plasma cells. Analogous to their normal B-cell counterparts, CD20 is expressed on malignant lymphoplasmacytic cells from most patients with Waldenstrom's macroglobulinemia and on malignant plasma cells from a fraction (20%) of multiple myeloma patients. CD20 also is expressed on subpopulations of normal donor plasma cells, which may include autoantibody-secreting plasmacytes. In view of these findings, the anti-CD20 chimeric monoclonal antibody, rituximab (Rituxan; Genentech, Inc, South San Francisco, CA and IDEC Pharmaceutical Corporation, San Diego, CA), has been evaluated in the treatment of Waldenstrom's macroglobulinemia and multiple myeloma, as well as in nonmalignant plasma cell disorders including IgM polyneuropathies, immune thrombocytopenias, and autoimmune hemolytic anemias, with reported activity in these entities. An update of these clinical efforts is presented in this report.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Paraproteinemias/tratamiento farmacológico , Anticuerpos Monoclonales de Origen Murino , Antígenos CD20/genética , Linfocitos B/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulina M/inmunología , Mieloma Múltiple/tratamiento farmacológico , Paraproteinemias/clasificación , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Rituximab , Factores de Transcripción , Macroglobulinemia de Waldenström/tratamiento farmacológico
19.
Semin Oncol ; 27(5): 598-613, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11049026

RESUMEN

The use of immunotherapy to treat patients with plasma cell dyscrasias (PCD) such as multiple myeloma (MM) and Waldenström's macroglobulinemia (WM) has gained enormous interest in recent years, with considerable efforts being mounted by many investigators. These efforts have included the use of serotherapy (antibody-mediated immunotherapy), vaccination strategies aimed at inducing allogeneic as well as autologous anti-MM immunity, and the use of donor lymphocyte infusions (DLIs). A number of cell surface antigens on malignant plasma cells and/or B cells in MM and/or WM patients have been proposed for use in tumor cell-targeted serotherapy, including immunoglobulin idiotype, CD19, CD20, CD38, CD54, CD138, HM1.24, and MUC1 core protein. Ongoing clinical trials are examining serotherapy targeting CD20 (in MM and WM) and CD38 (in MM), with early reports of responses to the anti-CD20 monoclonal antibody (mAb) Rituximab (Genentech, South San Francisco, CA) in patients with WM and certain patients with MM. The use of agents to induce MM- and WM-selective antigens for targeting in serotherapy has been proposed based on studies demonstrating the upregulation of CD20 by interferon-gamma (IFN-gamma), and of MUC1 core protein by dexamethasone (DEX) on malignant plasma cells. Strategies to induce allogeneic anti-MM immunity have included immunization of the marrow donor to idiotypic protein, as well as DLI. In addition, proposed immunization strategies aimed at inducing autologous immunity include vaccination with dendritic cells pulsed with MM antigens, MM cell-dendritic cell fusions, carrier-linked idiotype protein, catalytic subunit of telomerase, or DNA encoding for single-chain variable fragments (scFv) linked to a carrier protein gene. Whole tumor vaccination strategies are also being examined and include the use of MM cells transfected and/or stimulated with cytokines, costimulatory molecules, or CD40 ligand. Finally, potential obstacles to the use of immunotherapy, including the presence of resistance antigens on MM and WM tumor cells, are discussed.


Asunto(s)
Inmunización Pasiva , Inmunoterapia Activa , Paraproteinemias/terapia , Vacunas contra el Cáncer , Ensayos Clínicos como Asunto , Humanos , Inmunización Pasiva/métodos , Inmunoterapia Activa/métodos , Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/terapia , Macroglobulinemia de Waldenström/terapia
20.
J Immunol Methods ; 235(1-2): 11-9, 2000 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-10675753

RESUMEN

We developed a simple and rapid method to enrich tumor cells within bone marrow (BM) aspirates from patients with multiple myeloma (MM). Thirty patients with a median of 50% (8-85%) MM cells by morphology and 55% (6--85%) MM cells identified by CD38+CD45-cell surface phenotype were studied. BM mononuclear cells (BMMCs) were isolated by Ficoll Hypaque sedimentation and incubated with a cocktail of mouse monoclonal antibodies (mAbs) directed against CD3 (T cells); CD11b and CD14 (monocytes); CD33 (myeloid cells), CD45 and CD45RA (leucocyte common antigen); CD32 as well as glycophorin A. After the addition of anti-mouse Fc Ig-coated immunomagnetic beads, mAb-bound cells were removed in a magnetic field. The residual cell populations were enriched for MM cells, evidenced by >95% plasma cell morphology and >95% CD38+CD45RA-cell surface phenotype. Since this method requires only two short incubations, cell losses were minimal and the yield of MM cells was therefore high (>95%). Viability of the MM-cell enriched fractions was 99%, and these cells were functional in assays of proliferation, cell cycle analysis and immunoglobulin secretion. This immunomagnetic bead depletion method therefore permits the ready isolation of homogeneous populations of patient MM cells for use in both cellular and molecular studies.


Asunto(s)
Células de la Médula Ósea/patología , Técnicas de Cultivo de Célula/métodos , Separación Inmunomagnética , Mieloma Múltiple/patología , Supervivencia Celular , Humanos , Fenotipo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA