Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 127(11): 4042-4058, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972539

RESUMEN

NK cells, lymphocytes of the innate immune system, are important for defense against infectious pathogens and cancer. Classically, the CD56dim NK cell subset is thought to mediate antitumor responses, whereas the CD56bright subset is involved in immunomodulation. Here, we challenge this paradigm by demonstrating that brief priming with IL-15 markedly enhanced the antitumor response of CD56bright NK cells. Priming improved multiple CD56bright cell functions: degranulation, cytotoxicity, and cytokine production. Primed CD56bright cells from leukemia patients demonstrated enhanced responses to autologous blasts in vitro, and primed CD56bright cells controlled leukemia cells in vivo in a murine xenograft model. Primed CD56bright cells from multiple myeloma (MM) patients displayed superior responses to autologous myeloma targets, and furthermore, CD56bright NK cells from MM patients primed with the IL-15 receptor agonist ALT-803 in vivo displayed enhanced ex vivo functional responses to MM targets. Effector mechanisms contributing to IL-15-based priming included improved cytotoxic protein expression, target cell conjugation, and LFA-1-, CD2-, and NKG2D-dependent activation of NK cells. Finally, IL-15 robustly stimulated the PI3K/Akt/mTOR and MEK/ERK pathways in CD56bright compared with CD56dim NK cells, and blockade of these pathways attenuated antitumor responses. These findings identify CD56bright NK cells as potent antitumor effectors that warrant further investigation as a cancer immunotherapy.


Asunto(s)
Interleucina-15/farmacología , Células Asesinas Naturales/fisiología , Leucemia Mieloide Aguda/terapia , Mieloma Múltiple/terapia , Animales , Antígeno CD56/metabolismo , Degranulación de la Célula , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Humanos , Inmunidad Innata , Factores Inmunológicos/farmacología , Inmunoterapia , Integrinas/fisiología , Células K562 , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Proteínas/farmacología , Proteínas Recombinantes de Fusión , Transducción de Señal
2.
Biol Blood Marrow Transplant ; 23(3): 398-404, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27894857

RESUMEN

Cytokine-induced memory-like natural killer (NK) cells differentiate after short-term preactivation with IL-12, IL-15, and IL-18 and display enhanced effector function in response to cytokines or tumor targets for weeks after the initial preactivation. Conventional NK cell function depends on a licensing signal, classically delivered by an inhibitory receptor engaging its cognate MHC class I ligand. How licensing status integrates with cytokine-induced memory-like NK cell responses is unknown. We investigated this interaction using killer cell immunoglobulin-like receptor- and HLA-genotyped primary human NK cells. Memory-like differentiation resulted in enhanced IFN-γ production triggered by leukemia targets or FcγRIIIa ligation within licensed NK cells, which exhibited the highest functionality of the NK cell subsets interrogated. IFN-γ production by unlicensed memory-like NK cells was also enhanced to a level comparable with that of licensed control NK cells. Mechanistically, differences in responses to FcγRIIIa-based triggering were not explained by alterations in key signaling intermediates, indicating that the underlying biology of memory-like NK cells is distinct from that of adaptive NK cells in human cytomegalovirus-positive individuals. Additionally, memory-like NK cells responded robustly to cytokine receptor restimulation with no impact of licensing status. These results demonstrate that both licensed and unlicensed memory-like NK cell populations have enhanced functionality, which may be translated to improve leukemia immunotherapy.


Asunto(s)
Citocinas/farmacología , Memoria Inmunológica/efectos de los fármacos , Células Asesinas Naturales/inmunología , Receptores de IgG/inmunología , Receptores KIR/inmunología , Diferenciación Celular/efectos de los fármacos , Antígenos HLA , Humanos , Inmunoterapia Adoptiva , Interferón gamma/biosíntesis , Leucemia/terapia , Activación de Linfocitos
3.
Sci Transl Med ; 8(357): 357ra123, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27655849

RESUMEN

Natural killer (NK) cells are an emerging cellular immunotherapy for patients with acute myeloid leukemia (AML); however, the best approach to maximize NK cell antileukemia potential is unclear. Cytokine-induced memory-like NK cells differentiate after a brief preactivation with interleukin-12 (IL-12), IL-15, and IL-18 and exhibit enhanced responses to cytokine or activating receptor restimulation for weeks to months after preactivation. We hypothesized that memory-like NK cells exhibit enhanced antileukemia functionality. We demonstrated that human memory-like NK cells have enhanced interferon-γ production and cytotoxicity against leukemia cell lines or primary human AML blasts in vitro. Using mass cytometry, we found that memory-like NK cell functional responses were triggered against primary AML blasts, regardless of killer cell immunoglobulin-like receptor (KIR) to KIR-ligand interactions. In addition, multidimensional analyses identified distinct phenotypes of control and memory-like NK cells from the same individuals. Human memory-like NK cells xenografted into mice substantially reduced AML burden in vivo and improved overall survival. In the context of a first-in-human phase 1 clinical trial, adoptively transferred memory-like NK cells proliferated and expanded in AML patients and demonstrated robust responses against leukemia targets. Clinical responses were observed in five of nine evaluable patients, including four complete remissions. Thus, harnessing cytokine-induced memory-like NK cell responses represents a promising translational immunotherapy approach for patients with AML.


Asunto(s)
Citocinas/farmacología , Memoria Inmunológica/efectos de los fármacos , Células Asesinas Naturales/inmunología , Leucemia Mieloide Aguda/inmunología , Traslado Adoptivo , Anciano , Animales , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Leucemia Mieloide Aguda/patología , Ligandos , Masculino , Ratones , Persona de Mediana Edad , Receptores de Células Asesinas Naturales/metabolismo , Inducción de Remisión , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Cancer Res ; 22(3): 596-608, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26423796

RESUMEN

PURPOSE: Anti-CD20 monoclonal antibodies (mAb) are an important immunotherapy for B-cell lymphoma, and provide evidence that the immune system may be harnessed as an effective lymphoma treatment approach. ALT-803 is a superagonist IL-15 mutant and IL-15Rα-Fc fusion complex that activates the IL-15 receptor constitutively expressed on natural killer (NK) cells. We hypothesized that ALT-803 would enhance anti-CD20 mAb-directed NK-cell responses and antibody-dependent cellular cytotoxicity (ADCC). EXPERIMENTAL DESIGN: We tested this hypothesis by adding ALT-803 immunostimulation to anti-CD20 mAb triggering of NK cells in vitro and in vivo. Cell lines and primary human lymphoma cells were utilized as targets for primary human NK cells. Two complementary in vivo mouse models were used, which included human NK-cell xenografts in NOD/SCID-γc (-/-) mice. RESULTS: We demonstrate that short-term ALT-803 stimulation significantly increased degranulation, IFNγ production, and ADCC by human NK cells against B-cell lymphoma cell lines or primary follicular lymphoma cells. ALT-803 augmented cytotoxicity and the expression of granzyme B and perforin, providing one potential mechanism for this enhanced functionality. Moreover, in two distinct in vivo B-cell lymphoma models, the addition of ALT-803 to anti-CD20 mAb therapy resulted in significantly reduced tumor cell burden and increased survival. Long-term ALT-803 stimulation of human NK cells induced proliferation and NK-cell subset changes with preserved ADCC. CONCLUSIONS: ALT-803 represents a novel immunostimulatory drug that enhances NK-cell antilymphoma responses in vitro and in vivo, thereby supporting the clinical investigation of ALT-803 plus anti-CD20 mAbs in patients with indolent B-cell lymphoma.


Asunto(s)
Antineoplásicos/farmacología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Linfoma de Células B/inmunología , Linfoma de Células B/metabolismo , Proteínas/farmacología , Receptores de IgG/metabolismo , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Granzimas/genética , Granzimas/metabolismo , Humanos , Interferón gamma/biosíntesis , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Perforina/genética , Perforina/metabolismo , Proteínas Recombinantes de Fusión , Rituximab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Immunol ; 195(6): 2806-17, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268657

RESUMEN

NK cells develop in the bone marrow and complete their maturation in peripheral organs, but the molecular events controlling maturation are incompletely understood. The miR-15/16 family of microRNA regulates key cellular processes and is abundantly expressed in NK cells. In this study, we identify a critical role for miR-15/16 in the normal maturation of NK cells using a mouse model of NK-specific deletion, in which immature NK cells accumulate in the absence of miR-15/16. The transcription factor c-Myb (Myb) is expressed preferentially by immature NK cells, is a direct target of miR-15/16, and is increased in 15a/16-1 floxed knockout NK cells. Importantly, maturation of 15a/16-1 floxed knockout NK cells was rescued by Myb knockdown. Moreover, Myb overexpression in wild-type NK cells caused a defective NK cell maturation phenotype similar to deletion of miR-15/16, and Myb overexpression enforces an immature NK cell transcriptional profile. Thus, miR-15/16 regulation of Myb controls the NK cell maturation program.


Asunto(s)
Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , MicroARNs/genética , Proteínas Proto-Oncogénicas c-myb/genética , Regiones no Traducidas 3' , Traslado Adoptivo , Animales , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Células HEK293 , Humanos , Interferón gamma/biosíntesis , Células Asesinas Naturales/trasplante , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño
6.
Proc Natl Acad Sci U S A ; 112(7): E700-9, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25646418

RESUMEN

Phosphatase and tensin homolog (PTEN) is a critical negative regulator of the phosphoinositide-3 kinase pathway, members of which play integral roles in natural killer (NK) cell development and function. However, the functions of PTEN in NK cell biology remain unknown. Here, we used an NK cell-specific PTEN-deletion mouse model to define the ramifications of intrinsic NK cell PTEN loss in vivo. In these mice, there was a significant defect in NK cell numbers in the bone marrow and peripheral organs despite increased proliferation and intact peripheral NK cell maturation. Unexpectedly, we observed a significant expansion of peripheral blood NK cells and the premature egress of NK cells from the bone marrow. The altered trafficking of NK cells from peripheral organs into the blood was due to selective hyperresponsiveness to the blood localizing chemokine S1P. To address the importance of this trafficking defect to NK cell immune responses, we investigated the ability of PTEN-deficient NK cells to traffic to a site of tumor challenge. PTEN-deficient NK cells were defective at migrating to distal tumor sites but were more effective at clearing tumors actively introduced into the peripheral blood. Collectively, these data identify PTEN as an essential regulator of NK cell localization in vivo during both homeostasis and malignancy.


Asunto(s)
Movimiento Celular , Células Asesinas Naturales/inmunología , Fosfohidrolasa PTEN/fisiología , Animales , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA