Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 106(12): 3176-3187, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33147936

RESUMEN

Multiple myeloma (MM) disease progression is dependent on the ability of MM plasma cells (PCs) to egress from the bone marrow (BM), enter the circulation and disseminate to distal BM sites. Expression of the chemokine CXCL12 by BM stromal cells is crucial for MM PC retention within the BM. However, the mechanisms which overcome CXCL12-mediated retention to enable dissemination are poorly understood. We have previously identified that treatment with the CCR1 ligand CCL3 inhibits the response to CXCL12 in MM cell lines, suggesting that CCL3/CCR1 signalling may enable egress of MM PC from the BM. Here, we demonstrated that CCR1 expression was an independent prognostic indicator in newly diagnosed MM patients. Furthermore, we showed that CCR1 is a crucial driver of dissemination in vivo, with CCR1 expression in the murine MM cell line 5TGM1 being associated with an increased incidence of bone and splenic disseminated tumours in C57BL/KaLwRij mice. Furthermore, we demonstrated that CCR1 knockout in the human myeloma cell line OPM2 resulted in a >95% reduction in circulating MM PC numbers and BM and splenic tumour dissemination following intratibial injection in NSG mice. Therapeutic targeting of CCR1 with the inhibitor CCX9588 significantly reduced OPM2 or RPMI-8226 dissemination in intratibial xenograft models. Collectively, our findings suggest a novel role for CCR1 as a critical driver of BM egress of MM PCs during tumour dissemination. Furthermore, these data suggest that CCR1 may represent a potential therapeutic target for the prevention of MM tumour dissemination.


Asunto(s)
Mieloma Múltiple , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos C57BL , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Células Plasmáticas , Receptores CCR1/genética
2.
Blood ; 125(17): 2649-55, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25762180

RESUMEN

Pacritinib (SB1518) is a Janus kinase 2 (JAK2), JAK2(V617F), and Fms-like tyrosine kinase 3 inhibitor that does not inhibit JAK1. It demonstrated a favorable safety profile with promising efficacy in phase 1 studies in patients with primary and secondary myelofibrosis (MF). This multicenter phase 2 study further characterized the safety and efficacy of pacritinib in the treatment of patients with MF. Eligible patients had clinical splenomegaly poorly controlled with standard therapies or were newly diagnosed with intermediate- or high-risk Lille score. Patients with any degree of cytopenia were eligible. Thirty-five patients were enrolled. At entry, 40% had hemoglobin <10 g/dL and 43% had platelets <100 000× 10(9)/L. Up to week 24, 8 of 26 evaluable patients (31%) achieved a ≥35% decrease in spleen volume determined by magnetic resonance imaging and 14 of 33 (42%) attained a ≥50% reduction in spleen size by physical examination. Median MF symptom improvement was ≥50% for all symptoms except fatigue. Grade 1 or 2 diarrhea (69%) and nausea (49%) were the most common treatment-emergent adverse events. The study drug was discontinued in 9 patients (26%) due to adverse events (4 severe). Pacritinib is an active agent in patients with MF, offering a potential treatment option for patients with preexisting anemia and thrombocytopenia. This trial was registered at www.clinicaltrials.gov as #NCT00745550.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/uso terapéutico , Janus Quinasa 2/antagonistas & inhibidores , Mielofibrosis Primaria/tratamiento farmacológico , Pirimidinas/uso terapéutico , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Hidrocarburos Aromáticos con Puentes/efectos adversos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/patología , Pirimidinas/efectos adversos , Bazo/efectos de los fármacos , Bazo/patología , Esplenomegalia/complicaciones , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/patología
3.
Am J Hematol ; 92(6): 508-514, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28247421

RESUMEN

RBC-transfusion dependency (RBC-TD) is an independent prognostic factor for poor overall survival (OS) in the WHO classification-based prognostic scoring system (WPSS) for MDS patients. However, WPSS did not include cytopenia, whereas revised International Prognostic Scoring System (IPSS-R) did not include RBC-TD. Thus, neither of these prognostic scoring systems incorporates both cytopenia and RBC-TD. We aimed to test whether RBC-TD adds prognostic value to the IPSS-R. We analyzed MDS patients not treated with disease-modifying therapy, and enrolled in SA-MDS Registry (derivation cohort; n = 295) and Dusseldorf registry (Germany; validation cohort; n = 113) using time-dependent Cox proportional regression and serial landmark analyses. In the derivation cohort, RBC-TD patients had inferior OS compared to RBC transfusion-independent (RBC-TI) patients (P < 0.0001) at 6- (18 vs. 64 months), 12- (24 vs. 71 months), and 24-months (40 vs. 87 months). In a Cox proportional regression analysis, RBC-TD was an independent adverse prognostic marker in addition to age, sex, and IPSS-R variables (P < 0.0001). A prognostic index (PI) was derived using these Cox-proportional regression model variables. In the validation cohort, this PI classified patients into four prognostic groups with significantly different OS (P < 0.001) as in the derivation cohort. In conclusion, multivariate analysis by Cox proportional hazards regression and serial landmark analyses clearly demonstrates that development of RBC-TD at any time during the course of MDS is associated with poor OS, independent of IPSS-R. This study demonstrates that dynamic assessment of RBC-TD provides additional prognostic value to IPSS-R and should be included in treatment decision algorithms for MDS patients.


Asunto(s)
Transfusión de Eritrocitos , Síndromes Mielodisplásicos/mortalidad , Síndromes Mielodisplásicos/terapia , Adulto , Anciano , Anciano de 80 o más Años , Australia , Causas de Muerte , Terapia Combinada , Manejo de la Enfermedad , Transfusión de Eritrocitos/métodos , Femenino , Alemania , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Pronóstico , Modelos de Riesgos Proporcionales , Sistema de Registros , Reproducibilidad de los Resultados , Resultado del Tratamiento , Adulto Joven
4.
Exp Cell Res ; 332(1): 24-38, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25637218

RESUMEN

BACKGROUND: Increased expression of the tetraspanin TSPAN7 has been observed in a number of cancers; however, it is unclear how TSPAN7 plays a role in cancer progression. METHODS: We investigated the expression of TSPAN7 in the haematological malignancy multiple myleoma (MM) and assessed the consequences of TSPAN7 expression in the adhesion, migration and growth of MM plasma cells (PC) in vitro and in bone marrow (BM) homing and tumour growth in vivo. Finally, we characterised the association of TSPAN7 with cell surface partner molecules in vitro. RESULTS: TSPAN7 was found to be highly expressed at the RNA and protein level in CD138(+) MM PC from approximately 50% of MM patients. TSPAN7 overexpression in the murine myeloma cell line 5TGM1 significantly reduced tumour burden in 5TGM1/KaLwRij mice 4 weeks after intravenous adminstration of 5TGM1 cells. While TSPAN7 overexpression did not affect cell proliferation in vitro, TSPAN7 increased 5TGM1 cell adhesion to BM stromal cells and transendothelial migration. In addition, TSPAN7 was found to associate with the molecular chaperone calnexin on the cell surface. CONCLUSION: These results suggest that elevated TSPAN7 may be associated with better outcomes for up to 50% of MM patients.


Asunto(s)
Mieloma Múltiple/metabolismo , Proteínas del Tejido Nervioso/genética , Tetraspaninas/genética , Animales , Calnexina/genética , Calnexina/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Cocultivo , Expresión Génica , Humanos , Ratones Endogámicos C57BL , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , Trasplante de Neoplasias , Proteínas del Tejido Nervioso/metabolismo , Modelos de Riesgos Proporcionales , Tetraspaninas/metabolismo , Regulación hacia Arriba
5.
Biol Blood Marrow Transplant ; 19(5): 760-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23298856

RESUMEN

Autologous hematopoietic cell transplantation (AHCT) as initial therapy of patients with multiple myeloma (MM) improves survival. However, data to support this approach for relapsed/progressive disease after initial AHCT (AHCT1) are limited. Using Center for International Blood and Marrow Transplant Research data, we report the outcomes of 187 patients who underwent a second AHCT (AHCT2) for the treatment of relapsed/progressive MM. Planned tandem AHCT was excluded. Median age at AHCT2 was 59 years (range, 28 to 72), and median patient follow-up was 47 months (range, 3 to 97). Nonrelapse mortality after AHCT2 was 2% at 1 year and 4% at 3 years. Median interval from AHCT1 to relapse/progression was 18 months, and median interval between transplantations was 32 months. After AHCT2, the incidence of relapse/progression at 1 and 3 years was 51% and 82%, respectively. At 3 years after AHCT2, progression-free survival was 13%, and overall survival was 46%. In multivariate analyses, those relapsing ≥36 months after AHCT1 had superior progression-free (P = .045) and overall survival (P = .019). Patients who underwent AHCT2 after 2004 had superior survival (P = .026). AHCT2 is safe and feasible for disease progression after AHCT1. In this retrospective study, individuals relapsing ≥36 months from AHCT1 derived greater benefit from AHCT2 compared with those with a shorter disease-free interval. Storage of an adequate graft before AHCT1 will ensure that the option of a second autologous transplantation is retained for patients with relapsed/progressive MM.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Mieloma Múltiple/cirugía , Terapia Recuperativa/métodos , Adulto , Anciano , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Resultado del Tratamiento
6.
Blood ; 118(17): 4530-40, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21832280

RESUMEN

Transplantation with 2-5 × 10(6) mobilized CD34(+)cells/kg body weight lowers transplantation costs and mortality. Mobilization is most commonly performed with recombinant human G-CSF with or without chemotherapy, but a proportion of patients/donors fail to mobilize sufficient cells. BM disease, prior treatment, and age are factors influencing mobilization, but genetics also contributes. Mobilization may fail because of the changes affecting the HSC/progenitor cell/BM niche integrity and chemotaxis. Poor mobilization affects patient outcome and increases resource use. Until recently increasing G-CSF dose and adding SCF have been used in poor mobilizers with limited success. However, plerixafor through its rapid direct blockage of the CXCR4/CXCL12 chemotaxis pathway and synergy with G-CSF and chemotherapy has become a new and important agent for mobilization. Its efficacy in upfront and failed mobilizers is well established. To maximize HSC harvest in poor mobilizers the clinician needs to optimize current mobilization protocols and to integrate novel agents such as plerixafor. These include when to mobilize in relation to chemotherapy, how to schedule and perform apheresis, how to identify poor mobilizers, and what are the criteria for preemptive and immediate salvage use of plerixafor.


Asunto(s)
Neoplasias Hematológicas/terapia , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas/métodos , Hemoglobinuria Paroxística/terapia , Acondicionamiento Pretrasplante/métodos , Anemia Aplásica , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Humanos , Modelos Biológicos , Insuficiencia del Tratamiento
7.
Blood ; 114(23): 4859-70, 2009 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-19805619

RESUMEN

Deregulated cell survival programs are a classic hallmark of cancer. We have previously identified a serine residue (Ser585) in the betac subunit of the granulocyte-macrophage colony-stimulating factor receptor that selectively and independently promotes cell survival. We now show that Ser585 phosphorylation is constitutive in 20 (87%) of 23 acute myeloid leukemia (AML) patient samples, indicating that this survival-only pathway is frequently deregulated in leukemia. We performed a global expression screen to identify gene targets of this survival pathway and report a 138-gene betac Ser585-regulated transcriptome. Pathway analysis defines a gene network enriched for PI3-kinase target genes and a cluster of genes involved in cancer and cell survival. We show that one such gene, osteopontin (OPN), is a functionally relevant target of the Ser585-survival pathway as shown by siRNA-mediated knockdown of OPN expression that induces cell death in both AML blasts and CD34(+)CD38(-)CD123(+) leukemic progenitors. Increased expression of OPN at diagnosis is associated with poor prognosis with multivariate analysis indicating that it is an independent predictor of overall patient survival in normal karyotype AML (n = 60; HR = 2.2; P = .01). These results delineate a novel cytokine-regulated Ser585/PI3-kinase signaling network that is deregulated in AML and identify OPN as a potential prognostic and therapeutic target.


Asunto(s)
Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide/genética , Proteínas de Neoplasias/fisiología , Osteopontina/fisiología , Adulto , Anciano , Supervivencia Celular , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Femenino , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/mortalidad , Leucemia Mieloide/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Osteopontina/biosíntesis , Osteopontina/genética , Fosfatidilinositol 3-Quinasas/fisiología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfoserina/metabolismo , Pronóstico , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , ARN Interferente Pequeño/farmacología , Transducción de Señal/genética , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología
8.
Stem Cells ; 27(9): 2293-300, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19544531

RESUMEN

Limited cell numbers in a unit restricts cord blood transplantation (CBT) in adults. We evaluated whether cotransplantation of placental mesenchymal stromal cells (MSCs) would enhance engraftment. Plastic adherent cells from placenta demonstrated typical characteristics of MSCs. In six individual experiments, 4 cohorts of 24 nonobese diabetic/severe combined immune deficient (NOD/SCID) mice were evaluated. Cohort 1 received 5 x 10(4) CD34+ cells from unit (U) one (SCBT); cohort 2 received 5 x 10(4) CD34+ cells from U1 + 4 x 10(4) MSCs (SCBT+MSCs); cohort 3 received 2.5 x 10(4) CD34+ cells from U1 + 2.5 x 10(4) CD34+ cells from U2 (double cord blood transplant [DCBT]); cohort 4 received 2.5 x 10(4) CD34+ cells from U1 + 2.5 x 10(4) CD34+ cells from U2 + 4 x 10(4) MSCs (DCBT+MSCs). Hematopoietic engraftment evaluated after 6 to 8 weeks, was similar in recipients of SCBT and DCBT. MSC cotransplantation demonstrated enhanced engraftment in DCBT (51.8 +/- 6.8% versus 14.9 +/- 6.5%; p = .04) with an increased trend in SCBT (48.7 +/- 7.7% versus 17.5 +/- 6.1%; p = .07). In DCBT, cotransplantation of placental MSCs reduced single cord dominance. Self-renewal capacity was assessed by serial transplantation in secondary recipients infused with engrafted human cells from primary mice transplanted with or without MSCs. In secondary transplant experiments, 13 of 17 evaluable mice engrafted at levels of 1% to 6.5%. Despite enhanced engraftment in primary mice, long-term engraftment capacity was unaltered with MSC cotransplantation. Imaging studies showed MSCs migrated to pelvic region and improved cord blood (CB) CD34+ homing. Cotransplantation of placental MSCs enhanced cord blood engraftment and may act by improving homing of CD34+ cells.


Asunto(s)
Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Placenta/citología , Células del Estroma/citología , Animales , Antígenos CD34/metabolismo , Células Cultivadas , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Femenino , Citometría de Flujo , Humanos , Cariotipificación , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Embarazo , Células del Estroma/metabolismo , Células del Estroma/fisiología
9.
Clin Cancer Res ; 14(12): 3881-8, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559609

RESUMEN

PURPOSE: The organic cation transporter OCT-1 mediates active transport of imatinib. We recently showed that low OCT-1 activity is a major contributor to suboptimal response in chronic myeloid leukemia (CML) patients treated with imatinib. The relevance of OCT-1 activity and efflux pumps in determining intracellular uptake and retention (IUR) of dasatinib was assessed. EXPERIMENTAL DESIGN: The effect of OCT inhibitors on [14C]dasatinib and [14C]imatinib IUR was compared using peripheral blood mononuclear cells from newly diagnosed CML patients. The role of efflux transporters was studied using ABCB1- and ABCG2-overexpressing cell lines and relevant inhibitors. RESULTS: Unlike imatinib, there was no significant difference in the dasatinib IUR at 37 degrees C and 4 degrees C (P = 0.8), and OCT-1 inhibitors including prazosin did not reduce dasatinib IUR significantly. In CML mononuclear cells, prazosin inhibitable IUR was significantly higher for imatinib than dasatinib (6.38 versus 1.48 ng/200,000 cells; P = 0.002; n = 11). Patients with high OCT-1 activity based on their imatinib uptake had IC50(dasatinib) values equivalent to patients with low OCT-1 activity. Dasatinib IUR was significantly lower in ABCB1-overexpressing cell lines compared with parental cell lines (P < 0.05). PSC833 (ABCB1 inhibitor) significantly increased the dasatinib IUR (P < 0.05) and reduced IC50(dasatinib) (from 100 to 8 nmol/L) in K562-DOX cell line. The ABCG2 inhibitor Ko143 significantly increased dasatinib IUR in ABCG2-overexpressing cell lines and reduced IC(50)(dasatinib). CONCLUSION: Unlike imatinib, dasatinib cellular uptake is not significantly affected by OCT-1 activity, so that expression and function of OCT-1 is unlikely to affect response to dasatinib. Dasatinib is a substrate of both efflux proteins, ABCB1 and ABCG2.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Tiazoles/farmacocinética , Tiazoles/uso terapéutico , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Benzamidas , Transporte Biológico/efectos de los fármacos , Dasatinib , Evaluación Preclínica de Medicamentos , Proteínas de Fusión bcr-abl/metabolismo , Células HL-60 , Humanos , Mesilato de Imatinib , Concentración 50 Inhibidora , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Ratones , Proteínas de Neoplasias/fisiología , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/metabolismo , Piperazinas/farmacocinética , Temperatura , Células Tumorales Cultivadas
10.
Nat Genet ; 51(4): 694-704, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30926971

RESUMEN

Acute erythroid leukemia (AEL) is a high-risk leukemia of poorly understood genetic basis, with controversy regarding diagnosis in the spectrum of myelodysplasia and myeloid leukemia. We compared genomic features of 159 childhood and adult AEL cases with non-AEL myeloid disorders and defined five age-related subgroups with distinct transcriptional profiles: adult, TP53 mutated; NPM1 mutated; KMT2A mutated/rearranged; adult, DDX41 mutated; and pediatric, NUP98 rearranged. Genomic features influenced outcome, with NPM1 mutations and HOXB9 overexpression being associated with a favorable prognosis and TP53, FLT3 or RB1 alterations associated with poor survival. Targetable signaling mutations were present in 45% of cases and included recurrent mutations of ALK and NTRK1, the latter of which drives erythroid leukemogenesis sensitive to TRK inhibition. This genomic landscape of AEL provides the framework for accurate diagnosis and risk stratification of this disease, and the rationale for testing targeted therapies in this high-risk leukemia.


Asunto(s)
Leucemia Eritroblástica Aguda/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Genómica/métodos , Proteínas de Homeodominio/genética , Humanos , Lactante , Recién Nacido , Masculino , Mutación/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Nucleares/genética , Nucleofosmina , Pronóstico , Proteína p53 Supresora de Tumor/genética , Adulto Joven , Tirosina Quinasa 3 Similar a fms/genética
11.
Biol Blood Marrow Transplant ; 14(10): 1118-1124, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18804041

RESUMEN

Relapse is the overwhelming cause of treatment failure after autologous transplantation for multiple myeloma (MM). For patients with a syngeneic donor, twin transplants provide a healthy graft that is free of myeloma. The relative impact of the graft on posttransplant relapse can be estimated by comparing risk of relapse after hematopoietic cell transplantation from genetically identical twins versus autotransplants because confounding differences in minor or major histocompatibility antigens are absent in the syngeneic transplant setting. Outcomes of 43 subjects who received twin transplants for MM were compared to 170 matched autotransplant recipients reported to the Center for International Blood and Marrow Transplant Research (CIBMTR). Multivariate analysis was performed by fitting a Cox model stratified on matched pairs. The matched transplant patients studied were similar with respect to subject-, disease-, and transplant-related characteristics. Cumulative incidence of relapse/progression was significantly lower, and progression-free survival (PFS) was significantly higher following twin transplants. In multivariate analysis, the probability of relapse/progression was lower in twins (relative risk [RR] = 0.49, 95% confidence interval [CI] 0.28-0.86, P = .011). Twin transplants have a significantly lower relapse risk than autotransplants in MM, suggesting that graft composition may impact outcomes following high-dose chemotherapy.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Mieloma Múltiple/terapia , Inmunología del Trasplante , Adulto , Anciano , Análisis de Varianza , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Trasplante Autólogo/inmunología , Trasplante Isogénico/inmunología , Gemelos Monocigóticos
12.
Biol Blood Marrow Transplant ; 14(10): 1134-1140, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18804043

RESUMEN

Nonsecretory myeloma (NSM) accounts for <5% of cases of multiple myeloma (MM). The outcome of these patients following autologous stem cell transplantation (ASCT) has not been evaluated in clinical trials. We compared the outcomes after ASCT for patients with NSM reported to the Center for International Blood and Marrow Transplant Research (CIBMTR) between 1989 and 2003, to a matched group of 438 patients (4 controls for each patient) with secretory myeloma (SM). The patients were matched using propensity scores calculated using age, Durie-Salmon stage, sensitivity to pretransplant therapy, time from diagnosis to transplant, and year of transplant. Disease characteristics were similar in both groups at diagnosis and at transplant except higher risk of anemia, hypoalbuminemia, and marrow plasmacytosis (in SM) and plasmacytoma (more in NSM). Cumulative incidence of treatment-related mortality (TRM), relapse, progression-free survival (PFS), and overall survival (OS) were similar between the groups. In multivariate analysis, based on a Cox model stratified on matched pairs and adjusted for covariates not considered in the propensity score, we found no difference in outcome between the NSM and SM groups. In this large cohort of patients undergoing ASCT, we found no difference in outcomes of patients with NSM compared to those with SM.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/estadística & datos numéricos , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Adulto , Análisis de Varianza , Humanos , Persona de Mediana Edad , Mieloma Múltiple/complicaciones , Mieloma Múltiple/mortalidad , Recurrencia , Estudios Retrospectivos , Análisis de Supervivencia , Trasplante Autólogo , Resultado del Tratamiento
13.
Clin Cancer Res ; 12(23): 6973-7, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145816

RESUMEN

PURPOSE: Multiple myeloma is an incurable hematologic malignancy characterized by increased bone marrow angiogenesis and extensive lytic bone disease. We have previously shown that elevated levels of stromal-derived factor-1alpha (SDF-1alpha) in peripheral blood plasma are associated with osteolysis in multiple myeloma patients. We have now examined whether SDF-1alpha levels also correlate with angiogenesis. EXPERIMENTAL DESIGN: We examined the contribution of multiple myeloma plasma cell-derived SDF-1alpha in the stimulation of in vitro angiogenesis using a tube formation assay. We also collected trephine and peripheral blood plasma samples from patients with multiple myeloma to analyze microvessel density and SDF-1alpha levels, respectively. RESULTS: We show that multiple myeloma plasma cell line-derived conditioned medium containing SDF-1alpha stimulates in vitro angiogenesis. In addition, in a large cohort of patients with multiple myeloma and its precursor condition monoclonal gammopathy of undetermined significance, we confirm previous findings that plasma cell burden correlates with both angiogenesis and plasma levels of SDF-1alpha. We now extend these observations and show the novel finding that peripheral blood plasma levels of SDF-1alpha positively correlate with the degree of bone marrow angiogenesis in multiple myeloma and monoclonal gammopathy of undetermined significance patients. CONCLUSIONS: High levels of SDF-1alpha produced by multiple myeloma plasma cells promote osteolysis and bone marrow angiogenesis. Therefore, we propose that inhibition of SDF-1alpha may be an effective mechanism by which angiogenesis and osteolysis can be reduced in multiple myeloma patients.


Asunto(s)
Quimiocina CXCL12/sangre , Mieloma Múltiple/sangre , Neovascularización Patológica/sangre , Adulto , Anciano , Anciano de 80 o más Años , Médula Ósea/irrigación sanguínea , Médula Ósea/patología , Línea Celular Tumoral , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/farmacología , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/diagnóstico , Oligopéptidos/farmacología , Paraproteinemias , Valor Predictivo de las Pruebas , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/sangre , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad
14.
Cancer Res ; 65(5): 1700-9, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15753365

RESUMEN

Multiple myeloma (MM) is an incurable plasma cell (PC) malignancy able to mediate massive destruction of the axial and craniofacial skeleton. The aim of this study was to investigate the role of the potent chemokine, stromal-derived factor-1alpha (SDF-1alpha) in the recruitment of osteoclast precursors to the bone marrow. Our studies show that MM PC produce significant levels of SDF-1alpha protein and exhibit elevated plasma levels of SDF-1alpha when compared with normal, age-matched subjects. The level of SDF-1alpha positively correlated with the presence of multiple radiological bone lesions in individuals with MM, suggesting a potential role for SDF-1alpha in osteoclast precursor recruitment and activation. To examine this further, peripheral blood-derived CD14+ osteoclast precursors were cultured in an in vitro osteoclast-potentiating culture system in the presence of recombinant human SDF-1alpha. Although failing to stimulate an increase in TRAP+, multinucleated osteoclast formation, our studies show that SDF-1alpha mediated a dramatic increase in both the number and the size of the resorption lacunae formed. The increased osteoclast motility and activation in response to SDF-1alpha was associated with an increase in the expression of a number of osteoclast activation-related genes, including RANKL, RANK, TRAP, MMP-9, CA-II, and Cathepsin K. Importantly, the small-molecule CXCR4-specific inhibitor, 4F-Benzoyl-TE14011 (T140), effectively blocked osteoclast formation stimulated by the myeloma cell line, RPMI-8226. Based on these findings, we believe that the synthesis of high levels of SDF-1alpha by MM PC may serve to recruit osteoclast precursors to local sites within the bone marrow and enhance their motility and bone-resorbing activity. Therefore, we propose that inhibition of the CXCR4-SDF-1alpha axis may provide an effective means of treatment for MM-induced osteolysis.


Asunto(s)
Enfermedades Óseas/sangre , Quimiocinas CXC/sangre , Mieloma Múltiple/sangre , Osteoclastos/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Enfermedades Óseas/genética , Enfermedades Óseas/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Estudios de Casos y Controles , Movimiento Celular , Quimiocina CXCL12 , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Osteoclastos/citología , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Proteínas Recombinantes/metabolismo
15.
Cancer Res ; 77(20): 5452-5463, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28855206

RESUMEN

Disease progression and relapse in multiple myeloma is dependent on the ability of the multiple myeloma plasma cells (PC) to reenter the circulation and disseminate throughout the bone marrow. Increased bone marrow hypoxia is associated with increased recirculation of multiple myeloma PCs. Accordingly, we hypothesized that during chronic hypoxia, activation of HIF-2α may overcome the bone marrow retention signal provided by stromal-derived CXCL12, thereby enabling dissemination of multiple myeloma PCs. Here we demonstrate that HIF-2α upregulates multiple myeloma PC CXCL12 expression, decreasing migration toward CXCL12 and reducing adhesion to mesenchymal stromal cells in vitro We also found that HIF-2α strongly induced expression of the chemokine receptor CCR1 in multiple myeloma PCs. CCR1 activation potently induces multiple myeloma PC migration toward CCL3 while abrogating the multiple myeloma PC migratory response to CXCL12. In addition, increased CCR1 expression by multiple myeloma PCs conferred poor prognosis in newly diagnosed multiple myeloma patients and was associated with an increase in circulating multiple myeloma PCs in these patients. Taken together, our results suggest a role for hypoxia-mediated CCR1 upregulation in driving the egress of multiple myeloma PCs from the bone marrow. Targeting CCR1 may represent a novel strategy to prevent dissemination and overt relapse in multiple myeloma. Cancer Res; 77(20); 5452-63. ©2017 AACR.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Quimiocina CXCL12/metabolismo , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Células Plasmáticas/patología , Receptores CCR1/metabolismo , Receptores CXCR4/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Células Plasmáticas/metabolismo , Células Tumorales Cultivadas
16.
Cancer Res ; 63(17): 5438-45, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500379

RESUMEN

Multiple myeloma (MM) is an incurable B-cell malignancy able to mediate massive destruction of the axial skeleton. The aim of this study was to examine the involvement of the tumor necrosis factor-ligand family member, receptor activator of nuclear factor-kappaB ligand (RANKL), and its naturally occurring antagonist, osteoprotegerin (OPG), in MM biology. Using flow cytometry and two independent anti-RANKL antibodies, we demonstrate RANKL expression in CD38(+++)CD45(+) and CD38(+++)CD45(-) myeloma plasma cell (MPC) subpopulations derived from patients with osteolytic MM. In addition, highly purified subpopulations of MPC express mRNA for both transmembrane and soluble RANKL isoforms but lack expression of OPG mRNA and protein. We also show that RANKL expressed by MPC is functional as in vitro coculture of CD38(+++)CD45(+) and CD38(+++)CD45(-) MPC subpopulations with peripheral blood mononuclear cells resulted in the formation of multinucleate, tartrate-resistant acid phosphatase-positive osteoclasts-like cells capable of forming typical resorption pits. Furthermore, high expression of membrane-associated RANKL by CD38(+++) MPC correlated with the presence of multiple radiological bone lesions in individuals with MM. Together, our data strongly suggest that RANKL expression by MPC confers on them the ability to participate directly in the formation of osteoclast in vivo and extends our knowledge of the involvement of RANKL and OPG in the osteolysis characteristic of this disease.


Asunto(s)
Proteínas Portadoras/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Mieloma Múltiple/metabolismo , Osteoclastos/patología , Osteólisis/metabolismo , Proteínas Portadoras/genética , Citometría de Flujo , Expresión Génica , Humanos , Glicoproteínas de Membrana/genética , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Osteólisis/diagnóstico por imagen , Osteólisis/genética , Osteólisis/patología , Células Plasmáticas/metabolismo , Ligando RANK , Radiografía , Receptor Activador del Factor Nuclear kappa-B , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Methods Mol Med ; 113: 245-56, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15968108

RESUMEN

Much of the morbidity and mortality associated with the plasma cell (PC) malignancy, multiple myeloma (MM), is owing to the severe osteolytic bone disease seen in patients with this disease. Although the molecular mechanisms responsible for osteolysis remain to be fully elucidated, it is clear from numerous studies that it is owing, in part, to an increase in osteoclastic bone resorption. Several known osteoclast (OC)-activating factors (OAFs) are produced by myeloma PCs (MPCs), or by stromal cells in response to MPCs and include interleukin-1beta (IL-1beta); tumor necrosis factor-alpha (TNF-alpha); IL-6; parathyroid hormone-related protein; macrophage inflammatory protein-1alpha; and, most recently, the TNF-ligand family member receptor activator of nuclear factor-kappaB ligand (RANKL). The identification and significance of any one of these myeloma-derived OAFs is dependent on robust and reliable assays that measure the de novo formation and activation of OCs. A number of in vitro assay systems have been described that examine the requirements for normal OC formation and are easily adaptable for examining which MM-derived OAF and to what extent it is responsible for the bone loss observed in individuals with myeloma. This chapter describes one such in vitro model system.


Asunto(s)
Mieloma Múltiple/patología , Osteoclastos/patología , Osteólisis , Antígenos CD/sangre , Enfermedades Óseas/etiología , Enfermedades Óseas/patología , Resorción Ósea/patología , Huesos/patología , Huesos/ultraestructura , Técnicas de Cultivo de Célula/métodos , Técnicas de Cocultivo , Humanos , Leucocitos Mononucleares/patología , Receptores de Lipopolisacáridos/sangre , Microscopía Electrónica de Rastreo , Células Plasmáticas/patología
18.
Cancer Chemother Pharmacol ; 50(3): 171-8, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12203098

RESUMEN

PURPOSE: E21R is a competitive inhibitor of GM-CSF. This is the initial clinical study to investigate the safety, toxicity and pharmacokinetics of escalating doses of E21R. PATIENTS AND METHODS: Cohorts of three patients received doses of 10, 30, 100, 300, 600 and 1000 micro g/kg per day given subcutaneously daily for 10 days. Eligible patients had solid tumours known to express GM-CSF receptors (breast, prostate, colon and lung cancer, and melanoma). No bone marrow involvement or concomitant steroids were permitted. A total of 22 patients received doses ranging from 10 to 1000 micro g/kg per day. There were 18 males and 4 females with a median age of 60 years (range 33 to 81 years). Eight patients had an ECOG performance status of 0, seven a performance status of 1, and seven a performance status of 2. There were ten patients with colon cancer, four with prostate cancer, three with lung cancer, three with melanoma and two with breast cancer. RESULTS: E21R was in general well tolerated and the maximum tolerated dose was not reached. The most severe toxicities were WHO grade 3 injection site erythema in one patient and grade 2 in two patients, grade 2 lethargy in three patients and grade 2 muscle aches and soreness, grade 2 joint pains and grade 2 thirst in one patient each. The primary pharmacokinetic parameters were dose-independent. Dose-dependent transient eosinophilia was noted from day 3. A fall in PSA levels was recorded in two patients with prostate cancer during their initial cycles of E21R, but they subsequently rose again. Serum from patients treated at 600 and 1000 micro g/kg per day antagonized GM-CSF-mediated TF-1 cell proliferation in vitro. CONCLUSION: E21R can be safely given at doses up to 1000 micro g/kg per day.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Proteínas Recombinantes , Adulto , Anciano , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Eritema/inducido químicamente , Fatiga/inducido químicamente , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacocinética , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Neoplasias/tratamiento farmacológico , Dolor/inducido químicamente
19.
Nat Genet ; 45(3): 242-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23334668

RESUMEN

The genetic basis of hypodiploid acute lymphoblastic leukemia (ALL), a subtype of ALL characterized by aneuploidy and poor outcome, is unknown. Genomic profiling of 124 hypodiploid ALL cases, including whole-genome and exome sequencing of 40 cases, identified two subtypes that differ in the severity of aneuploidy, transcriptional profiles and submicroscopic genetic alterations. Near-haploid ALL with 24-31 chromosomes harbor alterations targeting receptor tyrosine kinase signaling and Ras signaling (71%) and the lymphoid transcription factor gene IKZF3 (encoding AIOLOS; 13%). In contrast, low-hypodiploid ALL with 32-39 chromosomes are characterized by alterations in TP53 (91.2%) that are commonly present in nontumor cells, IKZF2 (encoding HELIOS; 53%) and RB1 (41%). Both near-haploid and low-hypodiploid leukemic cells show activation of Ras-signaling and phosphoinositide 3-kinase (PI3K)-signaling pathways and are sensitive to PI3K inhibitors, indicating that these drugs should be explored as a new therapeutic strategy for this aggressive form of leukemia.


Asunto(s)
Aneuploidia , Aberraciones Cromosómicas , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Haploidia , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Ratones , Datos de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Trasplante Heterólogo , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
20.
Leuk Res ; 36(1): 110-6, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21993314

RESUMEN

Krüppel-like factor 5 (KLF5) has been implicated as a tumor suppressor in various solid tumors such as breast and prostate, and recent studies have demonstrated a role for this protein in neutrophil differentiation of acute promyelocytic leukemia cells in response to ATRA. Here, we show that KLF5 expression increases during primary granulocyte differentiation and that expression of KLF5 is a requirement for granulocyte differentiation of 32D cells. In AML, we show that KLF5 mRNA expression levels are reduced in multiple French-American-British subtypes compared to normal controls, and also in leukemic stem cells relative to normal hematopoietic stem cells. We demonstrate that in selected AML cases, reduced expression is associated with hypermethylation of the KLF5 locus in the proximal promoter and/or intron 1, suggesting that this may represent a Class II genetic lesion in the development of AML.


Asunto(s)
Metilación de ADN , Silenciador del Gen , Granulocitos/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Mieloide Aguda/genética , Línea Celular Tumoral , Metilación de ADN/fisiología , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Silenciador del Gen/fisiología , Sitios Genéticos , Granulocitos/patología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Análisis por Micromatrices , Regiones Promotoras Genéticas/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA