Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Leukemia ; 32(4): 996-1002, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29158558

RESUMEN

Arginine methyltransferases critically regulate cellular homeostasis by modulating the functional outcome of their substrates. The protein arginine methyltransferase 5 (PRMT5) is an enzyme involved in growth and survival pathways promoting tumorigenesis. However, little is known about the biologic function of PRMT5 and its therapeutic potential in multiple myeloma (MM). In the present study, we identified and validated PRMT5 as a new therapeutic target in MM. PRMT5 is overexpressed in patient MM cells and associated with decreased progression-free survival and overall survival. Either genetic knockdown or pharmacological inhibition of PRMT5 with the inhibitor EPZ015666 significantly inhibited growth of both cell lines and patient MM cells. Furthermore, PRMT5 inhibition abrogated NF-κB signaling. Interestingly, mass spectrometry identified a tripartite motif-containing protein 21 TRIM21 as a new PRMT5-partner, and we delineated a TRIM21-dependent mechanism of NF-κB inhibition. Importantly, oral administration of EPZ015666 significantly decreased MM growth in a humanized murine model of MM. These data both demonstrate the oncogenic role and prognostic relevance of PRMT5 in MM pathogenesis, and provide the rationale for novel therapies targeting PRMT5 to improve patient outcome.


Asunto(s)
Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Humanos , Isoquinolinas/farmacología , FN-kappa B/metabolismo , Pronóstico , Pirimidinas/farmacología , Ribonucleoproteínas/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Leukemia ; 32(3): 752-764, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29089645

RESUMEN

X-box binding protein 1 (XBP1), CD138 (Syndecan-1) and CS1 (SLAMF7) are highly expressed antigens in cancers including multiple myeloma (MM). Here, we identify and characterize immunogenic HLA-A24 peptides derived from these antigens for potential vaccination therapy of HLA-A24+ patients with MM. The identified immunogenic HLA-A24-specific XBP1 unspliced (UN)185-193 (I S P W I L A V L), XBP1 spliced (SP)223-231 (V Y P E G P S S L), CD138265-273 (I F A V C L V G F) and CS1240-248 (L F V L G L F L W) peptides induced antigen-specific CTL with anti-MM activity in an HLA-A24 restricted manner. Furthermore, a cocktail containing the four HLA-A24 peptides evoked MM-specific CTL with distinct phenotypic profiles (CD28, CD40L, 41BB, CD38, CD69) and anti-tumor activities, evidenced by perforin upregulation, CD107a degranulation (cytotoxicity) and Th1-type cytokines (IFN-γ/IL-2/TNF-α) production in response to HLA-A24+ MM cells. The multipeptide-specific CTL included antigen-specific memory CD8+ T cells expressing both T-cell activation (CD38, CD69) and immune checkpoints antigens (CTLA, PD-1, LAG-3, TIM-3). These results provide the framework for a multipeptide vaccination therapy to induce tumor-specific CTL in HLA-A24-positive patients with myeloma and other cancers expressing these antigens.


Asunto(s)
ADP-Ribosil Ciclasa 1/inmunología , Antígeno HLA-A24/inmunología , Mieloma Múltiple/inmunología , Péptidos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Proteína 1 de Unión a la X-Box/inmunología , ADP-Ribosil Ciclasa 1/química , ADP-Ribosil Ciclasa 1/metabolismo , Secuencia de Aminoácidos , Biomarcadores , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Epítopos de Linfocito T/química , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Antígeno HLA-A24/genética , Antígeno HLA-A24/metabolismo , Humanos , Memoria Inmunológica , Péptidos y Proteínas de Señalización Intercelular , Activación de Linfocitos/inmunología , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Péptidos/química , Péptidos/metabolismo , Fenotipo , Unión Proteica , Linfocitos T Citotóxicos/metabolismo , Proteína 1 de Unión a la X-Box/química , Proteína 1 de Unión a la X-Box/metabolismo
4.
Leukemia ; 31(12): 2670-2677, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28490812

RESUMEN

Epigenetic signaling pathways are implicated in tumorigenesis and therefore histone deacetylases (HDACs) represent novel therapeutic targets for cancers, including multiple myeloma (MM). Although non-selective HDAC inhibitors show anti-MM activities, unfavorable side effects limit their clinical efficacy. Isoform- and/or class-selective HDAC inhibition offers the possibility to maintain clinical activity while avoiding adverse events attendant to broad non-selective HDAC inhibition. We have previously reported that HDAC3 inhibition, either by genetic knockdown or selective inhibitor BG45, abrogates MM cell proliferation. Here we show that knockdown of HDAC3, but not HDAC1 or HDAC2, as well as BG45, downregulate expression of DNA methyltransferase 1 (DNMT1) mediating MM cell proliferation. DNMT1 expression is regulated by c-Myc, and HDAC3 inhibition triggers degradation of c-Myc protein. Moreover, HDAC3 inhibition results in hyperacetylation of DNMT1, thereby reducing the stability of DNMT1 protein. Combined inhibition of HDAC3 and DNMT1 with BG45 and DNMT1 inhibitor 5-azacytidine (AZA), respectively, triggers synergistic downregulation of DNMT1, growth inhibition and apoptosis in both MM cell lines and patient MM cells. Efficacy of this combination treatment is confirmed in a murine xenograft MM model. Our results therefore provide the rationale for combination treatment using HDAC3 inhibitor with DNMT1 inhibitor to improve patient outcome in MM.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1/genética , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Acetilación , Animales , Apoptosis , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Modelos Biológicos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Leukemia ; 31(12): 2661-2669, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28487543

RESUMEN

Recent studies have delineated cancer-type-specific roles of histone 3 lysine 27 (H3K27) demethylase KDM6B/JMJD3 depending on its H3K27 demethylase activity. Here we show that KDM6B is expressed in multiple myeloma (MM) cells; and that shRNA-mediated knockdown and CRISPR-mediated knockout of KDM6B abrogate MM cell growth and survival. Tumor necrosis factor-α or bone marrow stromal cell culture supernatants induce KDM6B, which is blocked by IKKß inhibitor MLN120B, suggesting that KDM6B is regulated by NF-κB signaling in MM cells. RNA-seq and subsequent ChIP-qPCR analyses reveal that KDM6B is recruited to the loci of genes encoding components of MAPK signaling pathway including ELK1 and FOS, and upregulates expression of these genes without affecting H3K27 methylation level. Overexpression of catalytically inactive KDM6B activates expression of MAPK pathway-related genes, confirming its function independent of demethylase activity. We further demonstrate that downstream targets of KDM6B, ELK1 and FOS, confer MM cell growth. Our study therefore delineates KDM6B function that links NF-κB and MAPK signaling pathway mediating MM cell growth and survival, and validates KDM6B as a novel therapeutic target in MM.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/metabolismo , Sistema de Señalización de MAP Quinasas , Mieloma Múltiple/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Mieloma Múltiple/genética , Mieloma Múltiple/mortalidad , Mieloma Múltiple/patología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Interferencia de ARN , Transducción de Señal , Proteína Elk-1 con Dominio ets/metabolismo
6.
Blood Cancer J ; 5: e312, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25978432

RESUMEN

Immunomodulatory drugs (IMiDs) thalidomide, lenalidomide (Len) and pomalidomide trigger anti-tumor activities in multiple myeloma (MM) by targetting cereblon and thereby impacting IZF1/3, c-Myc and IRF4. Histone deacetylase inhibitors (HDACi) also downregulate c-Myc. We therefore determined whether IMiDs with HDACi trigger significant MM cell growth inhibition by inhibiting or downregulating c-Myc. Combination treatment of Len with non-selective HDACi suberoylanilide hydroxamic acid or class-I HDAC-selective inhibitor MS275 induces synergic cytotoxicity, associated with downregulation of c-Myc. Unexpectedly, we observed that decreased levels of cereblon (CRBN), a primary target protein of IMiDs, was triggered by these agents. Indeed, sequential treatment of MM cells with MS275 followed by Len shows less efficacy than simultaneous treatment with this combination. Importantly ACY1215, an HDAC6 inhibitor with minimal effects on class-I HDACs, together with Len induces synergistic MM cytotoxicity without alteration of CRBN expression. Our results showed that only modest class-I HDAC inhibition is able to induce synergistic MM cytotoxicity in combination with Len. These studies may provide the framework for utilizing HDACi in combination with Len to both avoid CRBN downregulation and enhance anti-MM activities.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de Histona Desacetilasas/administración & dosificación , Inmunomodulación , Mieloma Múltiple/tratamiento farmacológico , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Ácidos Hidroxámicos/administración & dosificación , Immunoblotting , Técnicas In Vitro , Lenalidomida , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Talidomida/administración & dosificación , Talidomida/análogos & derivados , Transfección , Vorinostat
7.
Leukemia ; 29(9): 1918-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25801913

RESUMEN

Histone deacetylase (HDAC) inhibitors have been extensively investigated as therapeutic agents in cancer. However, the biological role of class IIa HDACs (HDAC4, 5, 7 and 9) in cancer cells, including multiple myeloma (MM), remains unclear. Recent studies show HDAC4 interacts with activating transcription factor 4 (ATF4) and inhibits activation of endoplasmic reticulum (ER) stress-associated proapoptotic transcription factor C/EBP homologous protein (CHOP). In this study, we hypothesized that HDAC4 knockdown and/or inhibition could enhance apoptosis in MM cells under ER stress condition by upregulating ATF4, followed by CHOP. HDAC4 knockdown showed modest cell growth inhibition; however, it markedly enhanced cytotoxicity induced by either tunicamycin or carfilzomib (CFZ), associated with upregulating ATF4 and CHOP. For pharmacological inhibition of HDAC4, we employed a novel and selective class IIa HDAC inhibitor TMP269, alone and in combination with CFZ. As with HDAC4 knockdown, TMP269 significantly enhanced cytotoxicity induced by CFZ in MM cell lines, upregulating ATF4 and CHOP and inducing apoptosis. Conversely, enhanced cytotoxicity was abrogated by ATF4 knockdown, confirming that ATF4 has a pivotal role mediating cytotoxicity in this setting. These results provide the rationale for novel treatment strategies combining class IIa HDAC inhibitors with ER stressors, including proteasome inhibitors, to improve patient outcome in MM.


Asunto(s)
Estrés del Retículo Endoplásmico , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Mieloma Múltiple/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Interleucina-6/metabolismo , Isoenzimas , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Inhibidores de Proteasoma/farmacología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
9.
Leukemia ; 28(3): 680-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23913134

RESUMEN

Histone deacetylases (HDACs) represent novel molecular targets for the treatment of various types of cancers, including multiple myeloma (MM). Many HDAC inhibitors have already shown remarkable antitumor activities in the preclinical setting; however, their clinical utility is limited because of unfavorable toxicities associated with their broad range HDAC inhibitory effects. Isoform-selective HDAC inhibition may allow for MM cytotoxicity without attendant side effects. In this study, we demonstrated that HDAC3 knockdown and a small-molecule HDAC3 inhibitor BG45 trigger significant MM cell growth inhibition via apoptosis, evidenced by caspase and poly (ADP-ribose) polymerase cleavage. Importantly, HDAC3 inhibition downregulates phosphorylation (tyrosine 705 and serine 727) of signal transducers and activators of transcription 3 (STAT3). Neither interleukin-6 nor bone marrow stromal cells overcome this inhibitory effect of HDAC3 inhibition on phospho-STAT3 and MM cell growth. Moreover, HDAC3 inhibition also triggers hyperacetylation of STAT3, suggesting crosstalk signaling between phosphorylation and acetylation of STAT3. Importantly, inhibition of HDAC3, but not HDAC1 or 2, significantly enhances bortezomib-induced cytotoxicity. Finally, we confirm that BG45 alone and in combination with bortezomib trigger significant tumor growth inhibition in vivo in a murine xenograft model of human MM. Our results indicate that HDAC3 represents a promising therapeutic target, and validate a prototype novel HDAC3 inhibitor BG45 in MM.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/efectos de los fármacos , Mieloma Múltiple/enzimología , División Celular , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Humanos , Mieloma Múltiple/patología
11.
Leukemia ; 27(12): 2366-75, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23807770

RESUMEN

Small-molecule multi-targeted cyclin-dependent kinase (CDK) inhibitors (CDKIs) are of particular interest due to their potent antitumor activity independent of p53 gene alterations. P53 deletion is associated with a very poor prognosis in multiple myeloma (MM). In this regard, we tested the anti-MM activity of RGB-286638, an indenopyrazole-derived CDKI with Ki-nanomolar activity against transcriptional CDKs. We examined RGB-286638's mode-of-action in MM cell lines with wild-type (wt)-p53 and those expressing mutant p53. RGB-286638 treatment resulted in MM cytotoxicity in vitro associated with inhibition of MM tumor growth and prolonged survival in vivo. RGB-286638 displayed caspase-dependent apoptosis in both wt-p53 and mutant-p53 cells that was closely associated with the downregulation of RNA polymerase II phosphorylation and inhibition of transcription. RGB-286638 triggered p53 accumulation via nucleolar stress and loss of Mdm2, accompanied by induction of p53 DNA-binding activity. In addition, RGB-286638 mediated p53-independent activity, which was confirmed by cytotoxicity in p53-knockdown and p53-mutant cells. We also demonstrated downregulation of oncogenic miR-19, miR-92a-1 and miR-21. Our data provide the rationale for the development of transcriptional CDKIs as therapeutic agents, which activate p53 in competent cells, while circumventing p53 deficiency through alternative p53-independent cell death mechanisms in p53-mutant/deleted cells.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Mieloma Múltiple/patología , Pirazoles/farmacología , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/fisiología , Urea/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Humanos , Masculino , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Leukemia ; 25(7): 1174-81, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21403648

RESUMEN

Upregulation of cytokines and chemokines is a frequent finding in multiple myeloma (MM). CCL3 (also known as MIP-1α) is a pro-inflammatory chemokine, levels of which in the MM microenvironment correlate with osteolytic lesions and tumor burden. CCL3 and its receptors, CCR1 and CCR5, contribute to the development of bone disease in MM by supporting tumor growth and regulating osteoclast (OC) differentiation. In this study, we identify inhibition of osteoblast (OB) function as an additional pathogenic mechanism in CCL3-induced bone disease. MM-derived and exogenous CCL3 represses mineralization and osteocalcin production by primary human bone marrow stromal cells and HS27A cells. Our results suggest that CCL3 effects on OBs are mediated by ERK activation and subsequent downregulation of the osteogenic transcription factor osterix. CCR1 inhibition reduced ERK phosphorylation and restored both osterix and osteocalcin expression in the presence of CCL3. Finally, treating SCID-hu mice with a small molecule CCR1 inhibitor suggests an upregulation of osteocalcin expression along with OC downregulation. Our results show that CCL3, in addition to its known catabolic activity, reduces bone formation by inhibiting OB function, and therefore contributes to OB/OC uncoupling in MM.


Asunto(s)
Remodelación Ósea/fisiología , Calcificación Fisiológica/fisiología , Quimiocina CCL3/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Mieloma Múltiple/complicaciones , Proteínas de Neoplasias/fisiología , Osteoblastos/fisiología , Osteocalcina/biosíntesis , Osteogénesis/fisiología , Osteólisis/etiología , Animales , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral/metabolismo , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Osteocalcina/genética , Osteoclastos/fisiología , Osteólisis/metabolismo , Osteólisis/patología , Receptores CCR1/biosíntesis , Receptores CCR1/genética , Receptores CCR5/biosíntesis , Receptores CCR5/genética , Factor de Transcripción Sp7 , Células del Estroma/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
14.
Leukemia ; 25(1): 161-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21102427

RESUMEN

LBH589 is a novel pan-histone deacetylase (HDAC) inhibitor that has potent antitumor activity in multiple myeloma and other hematological malignancies. However, its impact on the immune system has not been defined. We here evaluated the effects of LBH589 on human myeloid dendritic cells (DCs) at clinically relevant concentrations. Exposure to LBH589 affected the surface molecule expression on immature and mature DCs, which was associated with DC maturation (CD83↓), antigen presentation (human leukocyte antigen-ABC↓) and T-cell co-stimulation (CD40↓ and CD86↑). LBH589 decreased both protein and polysaccharide antigen uptake capacities by DCs. Importantly, LBH589 impaired DC function to stimulate antigen-specific immune responses, resulting in the significant reduction of invariant natural killer T-cell (CD1d-restricted) and T-cell (major histocompatibility complex-restricted) activation in innate and adaptive immunity. LBH589 also significantly repressed the production of interleukin (IL)-6, IL-10, IL-12p70, IL-23 and tumor necrosis factor-α by Toll-like receptor (TLR)3 and TLR4-induced DC activation, indicating an important role of HDAC activity in immune regulation and inflammation. RelB, a component of the nuclear factor-κ B signaling pathway, was the key component regulated by HDAC inhibition in DCs. Together, our preclinical study demonstrates that LBH589 significantly impairs the phenotype and function of DCs, indicating a need for monitoring the immune status in patients receiving HDAC inhibitor therapy. It also provides a rationale to evaluate LBH589 activity for the treatment of inflammation.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Células Mieloides/efectos de los fármacos , Antígenos CD40/análisis , Citocinas/biosíntesis , Células Dendríticas/fisiología , Humanos , Indoles , Activación de Linfocitos/efectos de los fármacos , Células Mieloides/fisiología , FN-kappa B/efectos de los fármacos , FN-kappa B/fisiología , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/inmunología , Panobinostat , Fenotipo
15.
Oncogene ; 29(16): 2325-36, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20101221

RESUMEN

Dysregulated cell cycling is a universal hallmark of cancer and is often mediated by abnormal activation of cyclin-dependent kinases (CDKs) and their cyclin partners. Overexpression of individual complexes are reported in multiple myeloma (MM), making them attractive therapeutic targets. In this study, we investigate the preclinical activity of a novel small-molecule multi-CDK inhibitor, AT7519, in MM. We show the anti-MM activity of AT7519 displaying potent cytotoxicity and apoptosis; associated with in vivo tumor growth inhibition and prolonged survival. At the molecular level, AT7519 inhibited RNA polymerase II (RNA pol II) phosphorylation, a CDK9, 7 substrate, associated with decreased RNA synthesis confirmed by [(3)H] Uridine incorporation. In addition, AT7519 inhibited glycogen synthase kinase 3beta (GSK-3beta) phosphorylation; conversely pretreatment with a selective GSK-3 inhibitor and shRNA GSK-3beta knockdown restored MM survival, suggesting the involvement of GSK-3beta in AT7519-induced apoptosis. GSK-3beta activation was independent of RNA pol II dephosphorylation confirmed by alpha-amanitin, a specific RNA pol II inihibitor, showing potent inhibition of RNA pol II phosphorylation without corresponding effects on GSK-3beta phosphorylation. These results offer new insights into the crucial, yet controversial role of GSK-3beta in MM and show significant anti-MM activity of AT7519, providing the rationale for its clinical evaluation in MM.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Piperidinas/farmacología , Pirazoles/farmacología , ARN Polimerasa II/antagonistas & inhibidores , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones SCID , Mieloma Múltiple/patología
16.
Leukemia ; 23(12): 2222-32, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19741729

RESUMEN

Although outcomes for patients with multiple myeloma (MM) have improved over the past decade, the disease remains incurable and even patients who respond well to induction therapy ultimately relapse and require additional treatment. Conventional chemotherapy and high-dose therapy with stem cell transplantation (SCT) have historically been utilized in the management of relapsed MM, but in recent years the immunomodulatory drugs (IMiDs) thalidomide and lenalidomide, as well as the proteasome inhibitor bortezomib, have assumed a primary role in this setting. This review focuses on the role of thalidomide, lenalidomide and bortezomib in relapsed and refractory MM, with additional discussion dedicated to emerging drugs in relapsed MM that may prove beneficial to patients with this disease.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Terapia Recuperativa/métodos , Antineoplásicos/uso terapéutico , Ácidos Borónicos/uso terapéutico , Bortezomib , Humanos , Inmunosupresores/uso terapéutico , Lenalidomida , Pirazinas/uso terapéutico , Talidomida/análogos & derivados , Talidomida/uso terapéutico , Resultado del Tratamiento
17.
Leukemia ; 23(9): 1679-86, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19369963

RESUMEN

Earlier studies have shown that ascorbic acid (vitamin C) inhibits bortezomib-induced cytotoxicity against cancer cells in vitro. However, the clinical significance of vitamin C on bortezomib treatment is unclear. In this study, we examined whether daily oral intake of vitamin C inhibits antimultiple myeloma (MM) activities of bortezomib. Vitamin C, at orally achievable concentrations, inhibited in vitro MM cell cytotoxicity of bortezomib and blocked its inhibitory effect on 20S proteasome activity. Specifically, plasma collected from healthy volunteers taking 1 g/day vitamin C reduced bortezomib-induced MM cell death in vitro. This antagonistic effect of vitamin C against proteasome inhibitors is limited to the boronate class of inhibitors (bortezomib and MG262). In vivo activity of this combination treatment was then evaluated using our xenograft model of human MM in SCID (severe combined immune-deficient) mice. Bortezomib (0.1 mg/kg twice a week for 4 weeks) significantly inhibits in vivo MM cell growth, which was blocked by oral vitamin C (40 mg/kg/day). Therefore, our results for the first time show that vitamin C can significantly reduce the activity of bortezomib treatment in vivo; and importantly, suggest that patients receiving treatment with bortezomib should avoid taking vitamin C dietary supplements.


Asunto(s)
Antineoplásicos/antagonistas & inhibidores , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Ácidos Borónicos/antagonistas & inhibidores , Pirazinas/antagonistas & inhibidores , Animales , Bortezomib , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma
18.
Leukemia ; 23(5): 961-70, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19151776

RESUMEN

Cyclin D dysregulation and overexpression is noted in the majority of multiple myeloma (MM) patients, suggesting its critical role in MM pathogenesis. Here, we sought to identify the effects of targeting cyclin D in MM. We first confirmed cyclin D mRNA overexpression in 42 of 64 (65%) patient plasma cells. Silencing cyclin D1 resulted in >50% apoptotic cell death suggesting its validity as a potential therapeutic target. We next evaluated P276-00, a clinical-grade small-molecule cyclin-dependent kinase inhibitor as a way to target the cyclins. P276-00 resulted in dose-dependent cytotoxicity in MM cells. Cell-cycle analysis confirmed either growth arrest or caspase-dependent apoptosis; this was preceded by inhibition of Rb-1 phosphorylation with associated downregulation of a range of cyclins suggesting a regulatory role of P276-00 in cell-cycle progression through broad activity. Proliferative stimuli such as interleukin-6, insulin-like growth factor-1 and bone-marrow stromal cell adherence induced cyclins; P276-00 overcame these growth, survival and drug resistance signals. Because the cyclins are substrates of proteasome degradation, combination studies with bortezomib resulted in synergism. Finally, in vivo efficacy of P276-00 was confirmed in an MM xenograft model. These studies form the basis of an ongoing phase I study in the treatment of relapsed/refractory MM.


Asunto(s)
Antineoplásicos/uso terapéutico , Ciclina D1/antagonistas & inhibidores , Flavonas/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Médula Ósea/efectos de los fármacos , Ácidos Borónicos/uso terapéutico , Bortezomib , Caspasas/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Regulación hacia Abajo , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Perfilación de la Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones SCID , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación/efectos de los fármacos , Pirazinas/uso terapéutico , Proteína de Retinoblastoma/metabolismo , Células del Estroma/efectos de los fármacos , Trasplante Heterólogo , Células Tumorales Cultivadas
19.
Leukemia ; 22(10): 1925-32, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18596740

RESUMEN

Osteolytic bone disease in multiple myeloma (MM) is caused by enhanced osteoclast (OCL) activation and inhibition of osteoblast function. Lenalidomide and bortezomib have shown promising response rates in relapsed and newly diagnosed MM, and bortezomib has recently been reported to inhibit OCLs. We here investigated the effect of lenalidomide on OCL formation and osteoclastogenesis in comparison with bortezomib. Both drugs decreased alpha V beta 3-integrin, tartrate-resistant acid phosphatase-positive cells and bone resorption on dentin disks. In addition, both agents decreased receptor activator of nuclear factor-kappaB ligand (RANKL) secretion of bone marrow stromal cells (BMSCs) derived from MM patients. We identified PU.1 and pERK as major targets of lenalidomide, and nuclear factor of activated T cells of bortezomib, resulting in inhibition of osteoclastogenesis. Furthermore, downregulation of cathepsin K, essential for resorption of the bone collagen matrix, was observed. We demonstrated a significant decrease of growth and survival factors including macrophage inflammatory protein-alpha, B-cell activating factor and a proliferation-inducing ligand. Importantly, in serum from MM patients treated with lenalidomide, the essential bone-remodeling factor RANKL, as well as the RANKL/OPG ratio, were significantly reduced, whereas osteoprotegerin (OPG) was increased. We conclude that both agents specifically target key factors in osteoclastogenesis, and could directly affect the MM-OCL-BMSCs activation loop in osteolytic bone disease.


Asunto(s)
Antineoplásicos/farmacología , Remodelación Ósea/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Osteoclastos/efectos de los fármacos , Talidomida/análogos & derivados , Factor Activador de Células B/metabolismo , Resorción Ósea/prevención & control , Ácidos Borónicos/farmacología , Bortezomib , Catepsina K , Catepsinas/análisis , Células Cultivadas , Quimiocina CCL3/metabolismo , Humanos , Integrina alfaVbeta3/análisis , Lenalidomida , Osteoclastos/fisiología , Osteoprotegerina/sangre , Proteínas Proto-Oncogénicas/fisiología , Pirazinas/farmacología , Ligando RANK/metabolismo , Talidomida/farmacología , Transactivadores/fisiología , Factor de Transcripción AP-1/fisiología , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
20.
Oncogene ; 27(6): 721-31, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-17653083

RESUMEN

Bortezomib is a proteasome inhibitor for the treatment of relapsed/refractory multiple myeloma (MM). Mechanisms of resistance to Bortezomib are undefined. Myeloid cell leukemia-1 (Mcl-1) is an antiapoptotic protein, which protects tumor cells against spontaneous and chemotherapy-induced apoptosis. In MM, specific downregulation of Mcl-1 induces apoptosis. Here, we examined the role of Mcl-1 in Bortezomib- and doxorubicin-induced apoptosis. We demonstrate that Bortezomib, but not doxorubicin, triggers caspase-dependent generation of a 28 kDa Mcl-1-fragment, in several MM cell lines, including MM.1S cells. Conversely, transient transfection of MM.1S cells with a previously reported 28 kDa Mcl-1(128-350) fragment, but not with the Mcl-1(1-127) fragment, induces apoptosis. Therefore, both downregulation of full-length antiapoptotic Mcl-1, as well as Bortezomib-induced generation of Mcl-1(128-350) cleaved protein, contribute to MM cell apoptosis. To verify further these findings, we next compared effects triggered by Bortezomib, doxorubicin and melphalan in Mcl-1(wt/wt) and Mcl-1(Delta/null) murine embryonic fibroblasts (MEFs). Our results show that Bortezomib, but not doxorubicin or melphalan, triggers Mcl-1 cleavage in Mcl-1(wt/wt), but not Mcl-1(Delta/null) MEFs and induces sub-G(1) phase cells; caspase-3 and -9, and PARP cleavage as well as morphological signs of apoptosis. Taken together, these results support an important role of Mcl-1 and a Mcl-1 fragment in Bortezomib-induced cell death in general, and in MM in particular. To prevent relapse of MM in patients treated with Bortezomib, we therefore recommend the combination of Bortezomib with agents that induce MM cell death independent of Mcl-1.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Ácidos Borónicos/farmacología , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirazinas/farmacología , Animales , Bortezomib , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos , Melfalán/farmacología , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...