Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Haematologica ; 97(9): 1348-56, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22419581

RESUMEN

BACKGROUND: Patients with gene expression profiling-defined high-risk myeloma in relapse have poor outcomes with current therapies. We tested whether natural killer cells expanded by co-culture with K562 cells transfected with 41BBL and membrane-bound interleukin-15 could kill myeloma cells with a high-risk gene expression profile in vitro and in a unique model which recapitulates human myeloma. DESIGN AND METHODS: OPM2 and high-risk primary myeloma tumors were grown in human fetal bone implanted into non-obese diabetic severe combined immunodeficiency mice with a deficient interleukin-2 receptor gamma chain. These mice are devoid of endogenous natural killer and T-cell activity and were used to determine whether adoptively transferred expanded natural killer cells could inhibit myeloma growth and myeloma-associated bone destruction. RESULTS: Natural killer cells from healthy donors and myeloma patients expanded a median of 804- and 351-fold, respectively, without significant T-cell expansion. Expanded natural killer cells killed both allogeneic and autologous primary myeloma cells avidly via a perforin-mediated mechanism in which the activating receptor NKG2D, natural cytotoxicity receptors, and DNAX-accessory molecule-1 played a central role. Adoptive transfer of expanded natural killer cells inhibited the growth of established OPM2 and high-risk primary myeloma tumors grown in the murine model. The transferred, expanded natural killer cells proliferated in vivo in an interleukin-2 dose-dependent fashion, persisted up to 4 weeks, were readily detectable in the human bone, inhibited myeloma growth and protected bone from myeloma-induced osteolysis. CONCLUSIONS: These studies provide the rationale for testing expanded natural killer cells in humans.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Mieloma Múltiple/terapia , Linfocitos T/inmunología , Animales , Apoptosis , Western Blotting , Proliferación Celular , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Osteólisis , Linfocitos T/metabolismo , Células Tumorales Cultivadas
2.
Cancer Res ; 65(21): 10041-9, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267030

RESUMEN

Myeloma vaccines, based on dendritic cells pulsed with idiotype or tumor lysate, have been met with limited success, probably in part due to insufficient cross-priming of myeloma antigens. A powerful method to introduce myeloma-associated antigens into the cytosol of dendritic cells is protein transduction, a process by which proteins fused with a protein transduction domain (PTD) freely traverse membrane barriers. NY-ESO-1, an immunogenic antigen by itself highly expressed in 60% of high-risk myeloma patients, was purified to near homogeneity both alone and as a recombinant fusion protein with a PTD, derived from HIV-Tat. Efficient entry of PTD-NY-ESO-1 into dendritic cells, confirmed by microscopy, Western blotting, and intracellular flow cytometry, was achieved without affecting dendritic cell phenotype. Experiments with amiloride, which inhibits endocytosis, and N-acetyl-l-leucinyl-l-norleucinal, a proteasome inhibitor, confirmed that PTD-NY-ESO-1 entered dendritic cells by protein transduction and was degraded by the proteasome. Tetramer analysis indicated superior generation of HLA-A2.1, CD8+ T lymphocytes specific for NY-ESO-1(157-165) with PTD-NY-ESO-1 compared with NY-ESO-1 control protein (44% versus 2%, respectively). NY-ESO-1-specific T lymphocytes generated with PTD-NY-ESO-1 secreted IFN-gamma indicative of a Tc1-type cytokine response. Thus, PTD-NY-ESO-1 accesses the cytoplasm by protein transduction, is processed by the proteasome, and NY-ESO-1 peptides presented by HLA class I elicit NY-ESO-1-specific T lymphocytes.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Proteínas de la Membrana/inmunología , Mieloma Múltiple/inmunología , Mieloma Múltiple/terapia , Amilorida/farmacología , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Células Dendríticas/metabolismo , Fibroblastos/fisiología , Antígenos HLA-A/inmunología , Antígeno HLA-A2 , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Monocitos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/inmunología , Transducción Genética
3.
Crit Rev Oncog ; 19(1-2): 121-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24941378

RESUMEN

Natural killer (NK) cells have been shown to have important functions in anti-tumor responses and therefore have been used as adoptive immunotherapy for cancer. Here, we review the current methods of ex vivo activation, enrichment, expansion, and shipment of clinical NK cell products.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Humanos
4.
PLoS One ; 8(1): e54610, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23372742

RESUMEN

Multicentric Castleman Disease is largely driven by increased signaling in the pathway for the plasma cell growth factor interleukin-6. We hypothesized that interleukin-6/interleukin-6 receptor/gp130 polymorphisms contribute to increased interleukin-6 and/or other components of the interleukin-6 signaling pathway in HIV-negative Castleman Disease patients. The study group was composed of 58 patients and 50 healthy donors of a similar racial/ethnic profile. Of seven polymorphisms chosen for analysis, we observed an increased frequency between patients and controls of the minor allele of interleukin-6 receptor polymorphism rs4537545, which is in linkage disequilibrium with interleukin-6 receptor polymorphism rs2228145. Further, individuals possessing at least one copy of the minor allele of either polymorphism expressed higher levels of soluble interleukin-6 receptor. These elevated interleukin-6 receptor levels may contribute to increased interleukin-6 activity through the trans-signaling pathway. These data suggest that interleukin-6 receptor polymorphism may be a contributing factor in Castleman Disease, and further research is warranted.


Asunto(s)
Enfermedad de Castleman/genética , Enfermedad de Castleman/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Alelos , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Interleucina-6/sangre , Interleucina-6/genética , Masculino
5.
Mol Cancer Ther ; 8(9): 2616-24, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19723891

RESUMEN

Monoclonal antibody (mAb) therapy for multiple myeloma, a malignancy of plasma cells, has not been clinically efficacious in part due to a lack of appropriate targets. We recently reported that the cell surface glycoprotein CS1 (CD2 subset 1, CRACC, SLAMF7, CD319) was highly and universally expressed on myeloma cells while having restricted expression in normal tissues. Elotuzumab (formerly known as HuLuc63), a humanized mAb targeting CS1, is currently in a phase I clinical trial in relapsed/refractory myeloma. In this report we investigated whether the activity of elotuzumab could be enhanced by bortezomib, a reversible proteasome inhibitor with significant activity in myeloma. We first showed that elotuzumab could induce patient-derived myeloma cell killing within the bone marrow microenvironment using a SCID-hu mouse model. We next showed that CS1 gene and cell surface protein expression persisted on myeloma patient-derived plasma cells collected after bortezomib administration. In vitro bortezomib pretreatment of myeloma targets significantly enhanced elotuzumab-mediated antibody-dependent cell-mediated cytotoxicity, both for OPM2 myeloma cells using natural killer or peripheral blood mononuclear cells from healthy donors and for primary myeloma cells using autologous natural killer effector cells. In an OPM2 myeloma xenograft model, elotuzumab in combination with bortezomib exhibited significantly enhanced in vivo antitumor activity. These findings provide the rationale for a clinical trial combining elotuzumab and bortezomib, which will test the hypothesis that combining both drugs would result in enhanced immune lysis of myeloma by elotuzumab and direct targeting of myeloma by bortezomib.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácidos Borónicos/uso terapéutico , Glicoproteínas de Membrana/inmunología , Mieloma Múltiple/tratamiento farmacológico , Pirazinas/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Apoptosis/efectos de los fármacos , Bortezomib , Estudios de Casos y Controles , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Glicoproteínas de Membrana/genética , Ratones , Ratones SCID , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Trasplante Heterólogo
6.
Blood ; 111(3): 1309-17, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17947507

RESUMEN

Human leukocyte antigen class I molecules expressed by tumor cells play a central role in the regulation of natural killer (NK) cell-mediated immune responses. The proteasome inhibitor bortezomib has demonstrated significant activity in multiple myeloma (MM). We hypothesized that treatment of MM with bortezomib results in the reduction of cell-surface expression of class I and thereby sensitizes MM to NK cell-mediated lysis. Here we report that bortezomib down-regulates class I in a time- and dose-dependent fashion on all MM cell lines and patient MM cells tested. Downregulation of class I can also be induced in vivo after a single dose of 1.0 mg/m(2) bortezomib. Bortezomib significantly enhances the sensitivity of patient myeloma to allogeneic and autologous NK cell-mediated lysis. Further, the level of decrease in class I expression correlates with increased susceptibility to lysis by NK cells. Clinically relevant bortezomib concentrations do not affect NK-cell function. Our findings have clear therapeutic implications for MM and other NK cell-sensitive malignancies in the context of both allogeneic and autologous adoptively transferred NK cells.


Asunto(s)
Ácidos Borónicos/farmacología , Membrana Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Mieloma Múltiple/inmunología , Pirazinas/farmacología , Bortezomib , Membrana Celular/inmunología , Supervivencia Celular/efectos de los fármacos , Humanos , Mieloma Múltiple/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Receptores KIR/clasificación , Receptores KIR/inmunología , Sensibilidad y Especificidad , Células Tumorales Cultivadas
7.
Blood ; 105(10): 3939-44, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15671442

RESUMEN

The presence of a metaphase cytogenetic abnormality (CA) is the key negative predictor of outcome in patients with multiple myeloma (MM). Gene expression profiling (GEP) of such patients showed increased expression of NY-ESO-1 compared to patients with normal cytogenetics (60% versus 31%; P = .004). NY-ESO-1 was also highly expressed in relapsing MM especially patients with CA (100% versus 60.7%; P < .001). GEP findings were confirmed at the protein level by immunostaining of marrow biopsies for NY-ESO-1. We detected spontaneous NY-ESO-1-specific antibodies by enzyme-linked immunosorbent assay in 33% of patients with NY-ESO-1+ MM, especially in CA patients (9 of 13; 70%), but in none of the NY-ESO-1- patients with MM (n = 27) or healthy donors (n = 21). Spontaneous NY-ESO-1(157-165)-specific T cells (0.2%-0.6% of CD8+ T cells) were found in the peripheral blood of NY-ESO-1+ MM with HLA-A*0201/NY-ESO-1(157-165) tetramers. These NY-ESO-1-specific T cells, when expanded, killed primary MM cells (50% lysis, effector-target [E/T] ratio, 10:1). Our data demonstrate that NY-ESO-1 is frequently expressed in MM with CA and is capable of eliciting spontaneous humoral and T-cell immunity. The pool of NY-ESO-1-specific cytotoxic T cells expands easily on NY-ESO-1 peptide stimulation and is functionally active. NY-ESO-1 should therefore be an ideal tumor target antigen for immunotherapy of patients with poor-prognosis MM.


Asunto(s)
Formación de Anticuerpos/inmunología , Antígenos de Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/metabolismo , Mieloma Múltiple/inmunología , Mieloma Múltiple/patología , Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Biopsia , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Humanos , Proteínas de la Membrana/inmunología , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/metabolismo , Pronóstico , Recurrencia , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA