Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
3.
Cancers (Basel) ; 13(11)2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34067236

RESUMEN

Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.

5.
Blood ; 136(2): 199-209, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32325491

RESUMEN

Granulocytic myeloid-derived suppressor cells (G-MDSCs) promote tumor growth and immunosuppression in multiple myeloma (MM). However, their phenotype is not well established for accurate monitoring or clinical translation. We aimed to provide the phenotypic profile of G-MDSCs based on their prognostic significance in MM, immunosuppressive potential, and molecular program. The preestablished phenotype of G-MDSCs was evaluated in bone marrow samples from controls and MM patients using multidimensional flow cytometry; surprisingly, we found that CD11b+CD14-CD15+CD33+HLADR- cells overlapped with common eosinophils and neutrophils, which were not expanded in MM patients. Therefore, we relied on automated clustering to unbiasedly identify all granulocytic subsets in the tumor microenvironment: basophils, eosinophils, and immature, intermediate, and mature neutrophils. In a series of 267 newly diagnosed MM patients (GEM2012MENOS65 trial), only the frequency of mature neutrophils at diagnosis was significantly associated with patient outcome, and a high mature neutrophil/T-cell ratio resulted in inferior progression-free survival (P < .001). Upon fluorescence-activated cell sorting of each neutrophil subset, T-cell proliferation decreased in the presence of mature neutrophils (0.5-fold; P = .016), and the cytotoxic potential of T cells engaged by a BCMA×CD3-bispecific antibody increased notably with the depletion of mature neutrophils (fourfold; P = .0007). Most interestingly, RNA sequencing of the 3 subsets revealed that G-MDSC-related genes were specifically upregulated in mature neutrophils from MM patients vs controls because of differential chromatin accessibility. Taken together, our results establish a correlation between the clinical significance, immunosuppressive potential, and transcriptional network of well-defined neutrophil subsets, providing for the first time a set of optimal markers (CD11b/CD13/CD16) for accurate monitoring of G-MDSCs in MM.


Asunto(s)
Antígenos CD , Mieloma Múltiple , Células Supresoras de Origen Mieloide , Proteínas de Neoplasias , Antígenos CD/sangre , Antígenos CD/genética , Antígenos CD/inmunología , Femenino , Estudios de Seguimiento , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/sangre , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Proteínas de Neoplasias/sangre , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Transcripción Genética/inmunología
6.
Leukemia ; 34(2): 589-603, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31595039

RESUMEN

The reason why a few myeloma cells egress from the bone marrow (BM) into peripheral blood (PB) remains unknown. Here, we investigated molecular hallmarks of circulating tumor cells (CTCs) to identify the events leading to myeloma trafficking into the bloodstream. After using next-generation flow to isolate matched CTCs and BM tumor cells from 32 patients, we found high correlation in gene expression at single-cell and bulk levels (r ≥ 0.94, P = 10-16), with only 55 genes differentially expressed between CTCs and BM tumor cells. CTCs overexpressed genes involved in inflammation, hypoxia, or epithelial-mesenchymal transition, whereas genes related with proliferation were downregulated in CTCs. The cancer stem cell marker CD44 was overexpressed in CTCs, and its knockdown significantly reduced migration of MM cells towards SDF1-α and their adhesion to fibronectin. Approximately half (29/55) of genes differentially expressed in CTCs were prognostic in patients with newly-diagnosed myeloma (n = 553; CoMMpass). In a multivariate analysis including the R-ISS, overexpression of CENPF and LGALS1 was significantly associated with inferior survival. Altogether, these results help understanding the presence of CTCs in PB and suggest that hypoxic BM niches together with a pro-inflammatory microenvironment induce an arrest in proliferation, forcing tumor cells to circulate in PB and seek other BM niches to continue growing.


Asunto(s)
Mieloma Múltiple/genética , Mieloma Múltiple/patología , Células Neoplásicas Circulantes/patología , Transcripción Genética/genética , Médula Ósea/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Expresión Génica/genética , Humanos , Hipoxia/genética , Hipoxia/patología , Inflamación/genética , Inflamación/patología , Células Madre Neoplásicas/patología , Pronóstico , Microambiente Tumoral/genética
7.
Clin Cancer Res ; 25(10): 3176-3187, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-30692097

RESUMEN

PURPOSE: Knowledge about the mechanism of action (MoA) of monoclonal antibodies (mAb) is required to understand which patients with multiple myeloma (MM) benefit the most from a given mAb, alone or in combination therapy. Although there is considerable research about daratumumab, knowledge about other anti-CD38 mAbs remains scarce. EXPERIMENTAL DESIGN: We performed a comprehensive analysis of the MoA of isatuximab. RESULTS: Isatuximab induces internalization of CD38 but not its significant release from MM cell surface. In addition, we uncovered an association between levels of CD38 expression and different MoA: (i) Isatuximab was unable to induce direct apoptosis on MM cells with CD38 levels closer to those in patients with MM, (ii) isatuximab sensitized CD38hi MM cells to bortezomib plus dexamethasone in the presence of stroma, (iii) antibody-dependent cellular cytotoxicity (ADCC) was triggered by CD38lo and CD38hi tumor plasma cells (PC), (iv) antibody-dependent cellular phagocytosis (ADCP) was triggered only by CD38hi MM cells, whereas (v) complement-dependent cytotoxicity could be triggered in less than half of the patient samples (those with elevated levels of CD38). Furthermore, we showed that isatuximab depletes CD38hi B-lymphocyte precursors and natural killer (NK) lymphocytes ex vivo-the latter through activation followed by exhaustion and eventually phagocytosis. CONCLUSIONS: This study provides a framework to understand response determinants in patients treated with isatuximab based on the number of MoA triggered by CD38 levels of expression, and for the design of effective combinations aimed at capitalizing disrupted tumor-stroma cell protection, augmenting NK lymphocyte-mediated ADCC, or facilitating ADCP in CD38lo MM patients.See related commentary by Malavasi and Faini, p. 2946.


Asunto(s)
Mieloma Múltiple/inmunología , ADP-Ribosil Ciclasa 1/inmunología , Anticuerpos Monoclonales , Anticuerpos Monoclonales Humanizados , Humanos
8.
Curr Osteoporos Rep ; 15(5): 499-506, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28889371

RESUMEN

PURPOSE OF THE REVIEW: Herein we dissect mechanisms behind the dissemination of cancer cells from primary tumor site to the bone marrow, which are necessary for metastasis development, with a specific focus on multiple myeloma. RECENT FINDINGS: The ability of tumor cells to invade vessels and reach the systemic circulation is a fundamental process for metastasis development; however, the interaction between clonal cells and the surrounding microenvironment is equally important for supporting colonization, survival, and growth in the secondary sites of dissemination. The intrinsic propensity of tumor cells to recognize a favorable milieu where to establish secondary growth is the basis of the "seed and soil" theory. This theory assumes that certain tumor cells (the "seeds") have a specific affinity for the milieu of certain organs (the "soil"). Recent literature has highlighted the important contributions of the vascular niche to the hospitable "soil" within the bone marrow. In this review, we discuss the crucial role of stromal cells and endothelial cells in supporting primary growth, homing, and metastasis to the bone marrow, in the context of multiple myeloma, a plasma cell malignancy with the unique propensity to primarily grow and metastasize to the bone marrow.


Asunto(s)
Médula Ósea/irrigación sanguínea , Neoplasias Óseas/secundario , Tejido Conectivo/irrigación sanguínea , Células Endoteliales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/patología , Médula Ósea/metabolismo , Tejido Conectivo/metabolismo , Células Endoteliales/citología , Humanos , Células Madre Mesenquimatosas/citología , Mieloma Múltiple/metabolismo , Metástasis de la Neoplasia , Microambiente Tumoral
9.
Oncoimmunology ; 6(4): e1283468, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28507790

RESUMEN

CD69 is an early activation marker on the surface of T lymphocytes undergoing activation by cognate antigen. We observed intense expression of CD69 on tumor-infiltrating T-lymphocytes that reside in the hypoxic tumor microenvironment and hypothesized that CD69 could be, at least partially, under the control of the transcriptional hypoxia response. In line with this, human and mouse CD3-stimulated lymphocytes cultured under hypoxia (1% O2) showed increased expression of CD69 at the protein and mRNA level. Consistent with these findings, mouse T lymphocytes that had recently undergone hypoxia in vivo, as denoted by pimonidazole staining, were more frequently CD69+ in the tumor and bone marrow hypoxic tissue compartments. We found evidence for HIF-1α involvement both when using T-lymphocytes from inducible HIF-1α-/- mice and when observing tumor-infiltrating T-lymphocytes in mice whose T cells are HIF-1α-/-. Direct pro-transcriptional activity of HIF-1α on a newly identified hypoxia response element (HRE) found in the human CD69 locus was demonstrated by ChIP experiments. These results uncover a connection between the HIF-1α oxygen-sensing pathway and CD69 immunobiology.

11.
Blood ; 127(15): 1896-906, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26755711

RESUMEN

Persistence of chemoresistant minimal residual disease (MRD) plasma cells (PCs) is associated with inferior survival in multiple myeloma (MM). Thus, characterization of the minor MRD subclone may represent a unique model to understand chemoresistance, but to our knowledge, the phenotypic and genetic features of the MRD subclone have never been investigated. Here, we compared the antigenic profile of MRD vs diagnostic clonal PCs in 40 elderly MM patients enrolled in the GEM2010MAS65 study and showed that the MRD subclone is enriched in cells overexpressing integrins (CD11a/CD11c/CD29/CD49d/CD49e), chemokine receptors (CXCR4), and adhesion molecules (CD44/CD54). Genetic profiling of MRD vs diagnostic PCs was performed in 12 patients; 3 of them showed identical copy number alterations (CNAs), in another 3 cases, MRD clonal PCs displayed all genetic alterations detected at diagnosis plus additional CNAs that emerged at the MRD stage, whereas in the remaining 6 patients, there were CNAs present at diagnosis that were undetectable in MRD clonal PCs, but also a selected number of genetic alterations that became apparent only at the MRD stage. The MRD subclone showed significant downregulation of genes related to protein processing in endoplasmic reticulum, as well as novel deregulated genes such as ALCAM that is prognostically relevant in MM and may identify chemoresistant PCs in vitro. Altogether, our results suggest that therapy-induced clonal selection could be already present at the MRD stage, where chemoresistant PCs show a singular phenotypic signature that may result from the persistence of clones with different genetic and gene expression profiles. This trial was registered atwww.clinicaltrials.gov as #NCT01237249.


Asunto(s)
Resistencia a Antineoplásicos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Neoplasia Residual/tratamiento farmacológico , Neoplasia Residual/genética , Anciano , Bortezomib/administración & dosificación , Moléculas de Adhesión Celular/metabolismo , Dexametasona/administración & dosificación , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Genómica , Humanos , Inmunofenotipificación , Integrinas/metabolismo , Lenalidomida , Masculino , Melfalán/administración & dosificación , Modelos Genéticos , Mieloma Múltiple/patología , Neoplasia Residual/patología , Fenotipo , Células Plasmáticas/patología , Prednisona/administración & dosificación , Pronóstico , Talidomida/administración & dosificación , Talidomida/análogos & derivados
12.
Cancer Res ; 75(10): 2071-82, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25769724

RESUMEN

Drug resistance remains a major clinical challenge for cancer treatment. Multiple myeloma is an incurable plasma cell cancer selectively localized in the bone marrow. The main cause of resistance in myeloma is the minimal residual disease cells that are resistant to the original therapy, including bortezomib treatment and high-dose melphalan in stem cell transplant. In this study, we demonstrate that altered tumor cell metabolism is essential for the regulation of drug resistance in multiple myeloma cells. We show the unprecedented role of the metabolic phenotype in inducing drug resistance through LDHA and HIF1A in multiple myeloma, and that specific inhibition of LDHA and HIF1A can restore sensitivity to therapeutic agents such as bortezomib and can also inhibit tumor growth induced by altered metabolism. Knockdown of LDHA can restore sensitivity of bortezomib resistance cell lines while gain-of-function studies using LDHA or HIF1A induced resistance in bortezomib-sensitive cell lines. Taken together, these data suggest that HIF1A and LDHA are important targets for hypoxia-driven drug resistance. Novel drugs that regulate metabolic pathways in multiple myeloma, specifically targeting LDHA, can be beneficial to inhibit tumor growth and overcome drug resistance.


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Resistencia a Antineoplásicos , Mieloma Múltiple/metabolismo , Pirazinas/farmacología , Animales , Bortezomib , Hipoxia de la Célula , Línea Celular Tumoral , Femenino , Glucólisis , Hexoquinasa/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Lactato Deshidrogenasa 5 , Ratones SCID , Mieloma Múltiple/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Blood ; 124(17): 2675-86, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25217697

RESUMEN

Proline-rich tyrosine kinase 2 (Pyk2) is a member of the focal adhesion kinase family that has been recently linked to tumor development. However, its role in modulating multiple myeloma (MM) biology and disease progression remains unexplored. We first demonstrated that patients with MM present with higher expression of Pyk2 compared with healthy individuals. By using loss-of-function approaches, we found that Pyk2 inhibition led to reduction of MM tumor growth in vivo as well as decreased cell proliferation, cell-cycle progression, and adhesion ability in vitro. In turn, overexpression of Pyk2 promoted the malignant phenotype, substantiated by enhanced tumor growth and reduced survival. Mechanistically, inhibition of Pyk2 reduced activation of Wnt/ß-catenin signaling by destabilizing ß-catenin, leading to downregulation of c-Myc and Cyclin D1. Furthermore, treatment of MM cells with the FAK/Pyk2 inhibitor VS-4718 effectively inhibited MM cell growth both in vitro and in vivo. Collectively, our findings describe the tumor-promoting role of Pyk2 in MM, thus providing molecular evidence for a novel tyrosine kinase inhibitor as a new therapeutic option in MM.


Asunto(s)
Aminopiridinas/farmacología , Quinasa 2 de Adhesión Focal/antagonistas & inhibidores , Mieloma Múltiple/prevención & control , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Progresión de la Enfermedad , Femenino , Quinasa 2 de Adhesión Focal/genética , Quinasa 2 de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Immunoblotting , Mediciones Luminiscentes , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
14.
Cell Rep ; 9(1): 118-128, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25263552

RESUMEN

Bone marrow (BM) metastasis remains one of the main causes of death associated with solid tumors as well as multiple myeloma (MM). Targeting the BM niche to prevent or modulate metastasis has not been successful to date. Here, we show that stromal cell-derived factor-1 (SDF-1/CXCL12) is highly expressed in active MM, as well as in BM sites of tumor metastasis and report on the discovery of the high-affinity anti-SDF-1 PEGylated mirror-image l-oligonucleotide (olaptesed-pegol). In vivo confocal imaging showed that SDF-1 levels are increased within MM cell-colonized BM areas. Using in vivo murine and xenograft mouse models, we document that in vivo SDF-1 neutralization within BM niches leads to a microenvironment that is less receptive for MM cells and reduces MM cell homing and growth, thereby inhibiting MM disease progression. Targeting of SDF-1 represents a valid strategy for preventing or disrupting colonization of the BM by MM cells.


Asunto(s)
Médula Ósea/patología , Quimiocina CXCL12/antagonistas & inhibidores , Mieloma Múltiple/terapia , Oligonucleótidos/farmacología , Animales , Médula Ósea/metabolismo , Neoplasias de la Médula Ósea/metabolismo , Neoplasias de la Médula Ósea/secundario , Ácidos Borónicos/farmacología , Bortezomib , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Metástasis de la Neoplasia , Oligonucleótidos/química , Oligonucleótidos/genética , Polietilenglicoles/química , Pirazinas/farmacología
15.
Blood ; 123(26): 4120-31, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24711662

RESUMEN

The C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in modulating cell trafficking in hematopoietic stem cells and clonal B cells. We screened 418 patients with B-cell lymphoproliferative disorders and described the presence of the C1013G/CXCR4 warts, hypogammaglobulinemia, infections, and myelokathexis-associated mutation in 28.2% (37/131) of patients with lymphoplasmacytic lymphoma (Waldenström macroglobulinemia [WM]), being either absent or present in only 7% of other B-cell lymphomas. In vivo functional characterization demonstrates its activating role in WM cells, as demonstrated by significant tumor proliferation and dissemination to extramedullary organs, leading to disease progression and decreased survival. The use of a monoclonal antibody anti-CXCR4 led to significant tumor reduction in a C1013G/CXCR4 WM model, whereas drug resistance was observed in mutated WM cells exposed to Bruton's tyrosine kinase, mammalian target of rapamycin, and phosphatidylinositol 3-kinase inhibitors, but not proteasome inhibitors. These findings demonstrate that C1013G/CXCR4 is an activating mutation in WM and support its role as a critical regulator of WM molecular pathogenesis and as an important therapeutic target.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Mutación Missense , Receptores CXCR4/metabolismo , Macroglobulinemia de Waldenström/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia sin Enfermedad , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones SCID , Metástasis de la Neoplasia , Trasplante de Neoplasias , Receptores CXCR4/genética , Tasa de Supervivencia , Macroglobulinemia de Waldenström/tratamiento farmacológico , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/mortalidad , Macroglobulinemia de Waldenström/patología
16.
PLoS One ; 8(6): e66982, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785514

RESUMEN

Glucocorticoid-induced TNF receptor (GITR) plays a crucial role in modulating immune response and inflammation, however the role of GITR in human cancers is poorly understood. In this study, we demonstrated that GITR is inactivated during tumor progression in Multiple Myeloma (MM) through promoter CpG island methylation, mediating gene silencing in primary MM plasma cells and MM cell lines. Restoration of GITR expression in GITR deficient MM cells led to inhibition of MM proliferation in vitro and in vivo and induction of apoptosis. These findings were supported by the presence of induction of p21 and PUMA, two direct downstream targets of p53, together with modulation of NF-κB in GITR-overexpressing MM cells. Moreover, the unbalanced expression of GITR in clonal plasma cells correlated with MM disease progression, poor prognosis and survival. These findings provide novel insights into the pivotal role of GITR in MM pathogenesis and disease progression.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Mieloma Múltiple/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Proliferación Celular , Islas de CpG , Metilación de ADN , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Humanos , Ratones , Modelos Biológicos , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , FN-kappa B/metabolismo , Pronóstico , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética
17.
Clin Lymphoma Myeloma Leuk ; 13(2): 205-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23474146

RESUMEN

Waldenström macroglobulinemia (WM) is a low-grade B-cell lymphoma characterized by the presence of lymphoplasmacytic cells in the BM (BM) and monoclonal immunoglobulin M in the circulation. Although WM cells showed minimal changes in cytogenetic studies and gene expression analysis, primary WM tumor cells present with a micro-RNA (miRNA) signature that differentiates them from their normal counterparts. This may suggest the importance of miRNAs in supporting WM pathogenesis. Among deregulated miRNAs, miRNA-155 has been shown to play a pivotal role in the biological characteristics of this disease both in vitro and in vivo, thus providing the rationale for testing miRNA-based therapeutic approaches for the treatment of WM.


Asunto(s)
Aberraciones Cromosómicas , MicroARNs/genética , Macroglobulinemia de Waldenström/genética , Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos
18.
J Clin Invest ; 123(4): 1542-55, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23454749

RESUMEN

BM mesenchymal stromal cells (BM-MSCs) support multiple myeloma (MM) cell growth, but little is known about the putative mechanisms by which the BM microenvironment plays an oncogenic role in this disease. Cell-cell communication is mediated by exosomes. In this study, we showed that MM BM-MSCs release exosomes that are transferred to MM cells, thereby resulting in modulation of tumor growth in vivo. Exosomal microRNA (miR) content differed between MM and normal BM-MSCs, with a lower content of the tumor suppressor miR-15a. In addition, MM BM-MSC-derived exosomes had higher levels of oncogenic proteins, cytokines, and adhesion molecules compared with exosomes from the cells of origin. Importantly, whereas MM BM-MSC-derived exosomes promoted MM tumor growth, normal BM-MSC exosomes inhibited the growth of MM cells. In summary, these in vitro and in vivo studies demonstrated that exosome transfer from BM-MSCs to clonal plasma cells represents a previously undescribed and unique mechanism that highlights the contribution of BM-MSCs to MM disease progression.


Asunto(s)
Neoplasias de la Médula Ósea/secundario , Exosomas/fisiología , Células Madre Mesenquimatosas/metabolismo , Mieloma Múltiple/patología , Anciano , Animales , Neoplasias de la Médula Ósea/metabolismo , Movimiento Celular , Proliferación Celular , Medios de Cultivo Condicionados , Progresión de la Enfermedad , Exosomas/metabolismo , Femenino , Fémur/patología , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Trasplante de Neoplasias , Andamios del Tejido , Carga Tumoral , Células Tumorales Cultivadas
19.
Clin Cancer Res ; 18(24): 6609-22, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23048077

RESUMEN

PURPOSE: The TORC1 inhibitor everolimus has previously shown significant activity as a single agent in hematologic malignancies, with reported responses of 30% to 70% in Waldenstrom macroglobulinemia. However, the specific mechanisms by which this class of mTOR inhibitors exerts anti-Waldenstrom macroglobulinemia activity have not been fully investigated. We therefore sought to dissect the mechanisms of everolimus-dependent modulation of Waldenstrom macroglobulinemia cell survival. EXPERIMENTAL DESIGN: We confirmed that everolimus targets mTOR in patients treated with everolimus and responding to therapy. We evaluated the effect of everolimus on proliferation and survival of primary Waldenstrom macroglobulinemia cells, as well as of other IgM-secreting lymphoma cell lines. Everolimus-dependent mechanisms of induced apoptosis and its effect on Waldenstrom macroglobulinemia cells in the context of bone marrow microenvironment have been also evaluated. miRNA-155 loss-of-function studies were conducted. Moreover, the combinatory effect of bortezomib and rituximab has been tested. RESULTS: We showed that everolimus targeted mTOR downstream signaling pathways, ex vivo, in patients responding to everolimus treatment. Everolimus induced toxicity in primary Waldenstrom macroglobulinemia cells, as well as in other IgM-secreting lymphoma cells, supported by cell-cycle arrest and caspase-dependent and -independent induction of apoptosis. Importantly, everolimus targeted Waldenstrom macroglobulinemia cells even in the context of bone marrow milieu, where it affected migration, adhesion, and angiogenesis. Everolimus-dependent anti-Waldenstrom macroglobulinemia activity was partially driven by miRNA-155. Moreover, everolimus synergized with bortezomib and rituximab in targeting Waldenstrom macroglobulinemia cells, as shown by synergistic inhibition of p65/ and p50/NF-κB activities. CONCLUSIONS: These findings provide a better understanding of the mechanisms that are responsible for everolimus-induced anti-Waldenstrom macroglobulinemia activity.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Complejos Multiproteicos/antagonistas & inhibidores , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Macroglobulinemia de Waldenström/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales de Origen Murino/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Médula Ósea/patología , Ácidos Borónicos/farmacología , Bortezomib , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos Clínicos Fase II como Asunto , Sinergismo Farmacológico , Everolimus , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , MicroARNs/fisiología , Pirazinas/farmacología , Rituximab , Transducción de Señal , Sirolimus/farmacología , Sirolimus/uso terapéutico , Resultado del Tratamiento , Células Tumorales Cultivadas/efectos de los fármacos , Microambiente Tumoral , Macroglobulinemia de Waldenström/patología
20.
Blood ; 120(8): 1678-86, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22797699

RESUMEN

miR-155 acts as an oncogenic miR in B-cell lymphoproliferative disorders, including Waldenstrom macroglobulinemia (WM) and chronic lymphocytic leukemia, and is therefore a potential target for therapeutic intervention. However, efficient targeting of miRs in tumor cells in vivo remains a significant challenge for the development of miR-155-based therapeutics for the treatment of B-cell malignancies. In the present study, we show that an 8-mer locked nucleic acid anti-miR-155 oligonucleotide targeting the seed region of miR-155 inhibits WM and chronic lymphocytic leukemia cell proliferation in vitro. Moreover, anti-miR-155 delivered systemically showed uptake in the BM CD19(+) cells of WM-engrafted mice, resulting in the up-regulation of several miR-155 target mRNAs in these cells, and decreased tumor growth significantly in vivo. We also found miR-155 levels to be elevated in stromal cells from WM patients compared with control samples. Interestingly, stromal cells from miR-155-knockout mice led to significant inhibition of WM tumor growth, indicating that miR-155 may also contribute to WM proliferation through BM microenvironmental cells. The results of the present study highlight the therapeutic potential of anti-miR-155-mediated inhibition of miR-155 in the treatment of WM.


Asunto(s)
Linfoma de Células B/genética , MicroARNs/genética , Oligonucleótidos Antisentido/uso terapéutico , Oligonucleótidos/uso terapéutico , Macroglobulinemia de Waldenström/genética , Animales , Proliferación Celular , Femenino , Silenciador del Gen , Terapia Genética , Humanos , Linfoma de Células B/terapia , Ratones , Ratones Endogámicos BALB C , Oligonucleótidos/genética , Oligonucleótidos Antisentido/genética , Células Tumorales Cultivadas , Macroglobulinemia de Waldenström/patología , Macroglobulinemia de Waldenström/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...