Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 418
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34404729

RESUMEN

Chimeric antigen receptor (CAR)-engineered T cell therapies have been recognized as powerful strategies in cancer immunotherapy; however, the clinical application of CAR-T is currently constrained by severe adverse effects in patients, caused by excessive cytotoxic activity and poor T cell control. Herein, we harnessed a dietary molecule resveratrol (RES)-responsive transactivator and a transrepressor to develop a repressible transgene expression (RESrep) device and an inducible transgene expression (RESind) device, respectively. After optimization, these tools enabled the control of CAR expression and CAR-mediated antitumor function in engineered human cells. We demonstrated that a resveratrol-repressible CAR expression (RESrep-CAR) device can effectively inhibit T cell activation upon resveratrol administration in primary T cells and a xenograft tumor mouse model. Additionally, we exhibit how a resveratrol-inducible CAR expression (RESind-CAR) device can achieve fine-tuned and reversible control over T cell activation via a resveratrol-titratable mechanism. Furthermore, our results revealed that the presence of RES can activate RESind-CAR T cells with strong anticancer cytotoxicity against cells in vitro and in vivo. Our study demonstrates the utility of RESrep and RESind devices as effective tools for transgene expression and illustrates the potential of RESrep-CAR and RESind-CAR devices to enhance patient safety in precision cancer immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Inmunoterapia Adoptiva/métodos , Leucemia Eritroblástica Aguda/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/terapia , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Immunol Methods ; 497: 113100, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34270976

RESUMEN

In recent years, adoptive cell therapy of immune effector cells, such as chimeric antigen receptor-T (CAR-T) cells, natural killer (NK) cells, and epitope-specific cytotoxic T lymphocyte (CTL) cells have been employed in clinical trials. In addition, CD19 CAR-T cells have been approved by the FDA for treatment of non-Hodgkin lymphoma and diffuse large B-cell lymphoma. In this context, it is vital to detect cellular cytotoxicity and monitor the quality of ex vivo expanded immune cells before product release and patient infusion. Target cells could proliferate in parallel with effector cells during the cytotoxicity assay, making it difficult to estimate the death ratio using conventional approaches. Meanwhile, non-specific dyes or non-homogeneous biomarkers for target cells may interfere with the final readout post addition of effector cells. Here, we modified a component of the coincubation medium to suppress the spontaneous release of bis(acetoxymethyl)2,2':6',2″-terpyridine-6,6″-dicarboxylate and sustained the window at a stable range (~70%). Further, the optimized Eu-TDA method presented reliable outcomes compared with lactate dehydrogenase detection and was compatible with cytotoxicity tests for NK cells and specific CTLs. Finally, the reported assay can accurately detect death of target cells depending on the amount of hydrophilic complex and can be reliably applied in quality control and cell activity evaluation tests on co-suspended effector and target cells. SUMMARY: A medium component for cellular coincubations (and associated protocols) have been optimized and validated for cytotoxicity assays, which can reliably evaluate the potency of engineered CD19 CAR-T cells, NK cells, and specific CTLs. In particular, the reported method can be applied widely in routine assays for bi-suspended effector and target cells with a stable window.


Asunto(s)
Citotoxicidad Inmunológica , Inmunohistoquímica , Inmunoterapia Adoptiva , Células Asesinas Naturales/trasplante , Leucemia Eritroblástica Aguda/terapia , Linfocitos T Citotóxicos/trasplante , Antígenos CD19/genética , Antígenos CD19/inmunología , Supervivencia Celular , Técnicas de Cocultivo , Epítopos , Citometría de Flujo , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/patología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T Citotóxicos/inmunología
3.
Curr Oncol Rep ; 23(6): 69, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33876292

RESUMEN

PURPOSE OF THE REVIEW: Acute erythroleukemia (AEL) is a rare form of acute myeloid leukemia recognized by erythroblastic proliferation. Many controversies remain around diagnosis influencing prognostic and therapeutic implications relating to this unique leukemia subset. RECENT FINDINGS: The 2016 WHO classification includes more clear and restrictive diagnostic criteria for AEL. Primary acute erythroid leukemia is associated with complex and high-risk karyotypes including chromosomes 5q and 7q abnormalities. Mutational data shows that AEL is characterized by far lower NPM1 and FLT3-ITD mutation rates and higher mutational rates in TP53 compared with other AML subtypes. Hypomethylating agents have shown therapeutic value in AEL. In this article, we discuss the evolving diagnostic concepts of erythroleukemia, genomics, clinical outcome, and promising therapeutic targets through an appraisal of the current literature.


Asunto(s)
Leucemia Eritroblástica Aguda/tratamiento farmacológico , Aberraciones Cromosómicas , Humanos , Inmunofenotipificación , Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Mutación
4.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33737343

RESUMEN

BACKGROUND: CAR-T cells immunotherapy is a breakthrough in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and B-cell malignancies. However, CAR-T therapies face major hurdles such as the lack of tumor-specific antigen (TSA), and immunosuppressive tumor microenvironment sometimes caused by the tumorous expression of immune checkpoints (ICPs) such as HLA-G. Indeed, HLA-G is remarkable because it is both a potent ICP and a TSA. HLA-G tumor expression causes immune escape by impairing innate and adaptive immune responses and by inducing a suppressive microenvironment. Yet, to date, no immunotherapy targets it. METHODS: We have developed two anti-HLA-G third-generation CARs based on new anti-HLA-G monoclonal antibodies. RESULTS: Anti-HLA-G CAR-T cells were specific for immunosuppressive HLA-G isoforms. HLA-G-activated CAR-T cells polarized toward T helper 1, and became cytotoxic against HLA-G+ tumor cells. In vivo, anti-HLA-G CAR-T cells were able to control and eliminate HLA-G+ tumor cells. The interaction of tumor-HLA-G with interleukin (IL)T2-expressing T cells is known to result in effector T cell functional inhibition, but anti-HLA-G CAR-T cells were insensitive to this inhibition and still exerted their function even when expressing ILT2. Lastly, we show that anti-HLA-G CAR-T cells differentiated into long-term memory effector cells, and seemed not to lose function even after repeated stimulation by HLA-G-expressing tumor cells. CONCLUSION: We report for the first time that HLA-G, which is both a TSA and an ICP, constitutes a valid target for CAR-T cell therapy to specifically target and eliminate both tumor cells and HLA-G+ suppressive cells.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos HLA-G/metabolismo , Inmunoterapia Adoptiva , Leucemia Eritroblástica Aguda/terapia , Células T de Memoria/trasplante , Receptores Quiméricos de Antígenos/genética , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Antígenos CD/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Antígenos HLA-G/inmunología , Humanos , Memoria Inmunológica , Células K562 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/metabolismo , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Receptores Quiméricos de Antígenos/metabolismo , Factores de Tiempo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
MAbs ; 11(3): 593-605, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30604643

RESUMEN

Targeting transferrin receptor 1 (TfR1) with monoclonal antibodies is a promising therapeutic strategy in cancer as tumor cells often overexpress TfR1 and show increased iron needs. We have re-engineered six anti-human TfR1 single-chain variable fragment (scFv) antibodies into fully human scFv2-Fcγ1 and IgG1 antibodies. We selected the more promising candidate (H7), based on its ability to inhibit TfR1-mediated iron-loaded transferrin internalization in Raji cells (B-cell lymphoma). The H7 antibody displayed nanomolar affinity for its target in both formats (scFv2-Fcγ1 and IgG1), but cross-reacted with mouse TfR1 only in the scFv2-Fc format. H7 reduced the intracellular labile iron pool and, contrary to what has been observed with previously described anti-TfR1 antibodies, upregulated TfR1 level in Raji cells. H7 scFv2-Fc format elimination half-life was similar in FcRn knock-out and wild type mice, suggesting that TfR1 recycling contributes to prevent H7 elimination in vivo. In vitro, H7 inhibited the growth of erythroleukemia and B-cell lymphoma cell lines (IC50 0.1 µg/mL) and induced their apoptosis. Moreover, the Im9 B-cell lymphoma cell line, which is resistant to apoptosis induced by rituximab (anti-CD20 antibody), was sensitive to H7. In vivo, tumor regression was observed in nude mice bearing ERY-1 erythroleukemia cell xenografts treated with H7 through a mechanism that involved iron deprivation and antibody-dependent cytotoxic effector functions. Therefore, targeting TfR1 using the fully human anti-TfR1 H7 is a promising tool for the treatment of leukemia and lymphoma.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/inmunología , Antineoplásicos Inmunológicos , Leucemia Eritroblástica Aguda , Receptores de Transferrina/inmunología , Anticuerpos de Cadena Única , Animales , Antineoplásicos Inmunológicos/inmunología , Antineoplásicos Inmunológicos/farmacología , Células CHO , Cricetulus , Células HEK293 , Humanos , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/patología , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacología
6.
Life Sci Alliance ; 3(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-32024667

RESUMEN

Studies of isolated cells, mice, and humans have demonstrated the vital role of the FERM domain protein kindlin-3 in integrin activation in certain hematopoietic and non-hematopoietic cells, consequent to binding to integrin ß-subunits. To explore regulatory mechanisms, we developed a monoclonal antibody that selectively recognizes the phosphorylated form of Ser484 (pS484) in kindlin-3. Activation of platelets, HEL megakaryocytic-like cells and BT549 breast cancer cells led to enhanced expression of pS484 as assessed by immunofluorescence or Western blotting. In platelets, pS484 rose rapidly and transiently upon stimulation. When a mutant form of kindlin-3, T482S484/AA kindlin-3, was transduced into mouse megakaryocytes, it failed to support activation of integrin αIIbß3, whereas wild-type kindlin-3 did. In MDA-MB231 breast cancer cells, expression of T482S484/AA kindlin-3 suppressed cell spreading, migration, invasion, and VEGF production. Wild-type kindlin-3 expressing cells markedly increased tumor growth in vivo, whereas T482S484/AA kindlin-3 significantly blunted tumor progression. Thus, our data establish that a unique phosphorylation event in kindlin-3 regulates its cellular functions.


Asunto(s)
Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Plaquetas/citología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Células CHO , Línea Celular Tumoral , Cricetulus , Proteínas del Citoesqueleto/inmunología , Proteínas del Citoesqueleto/metabolismo , Femenino , Humanos , Cadenas beta de Integrinas/metabolismo , Leucemia Eritroblástica Aguda/inmunología , Ratones , Ratones Desnudos , Fosforilación , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo
7.
JCI Insight ; 3(9)2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29720574

RESUMEN

Using a mouse retroviral model, we have shown that mAb-based immunotherapy can induce life-long endogenous protective immunity (vaccine-like effects). This observation has potentially important consequences for treating life-threatening human viral infections. Here, we investigated the role of neutrophils in this effect. Neutrophils are innate immunity effector cells with well-established microbe-killing activities that are rapidly mobilized upon infection. They are also emerging as orchestrators of innate and adaptive immunities. However, their immunomodulatory activity during antiviral mAb immunotherapies has never been studied. Our data reveal that neutrophils have an essential role in immunotherapy-induced immune protection of infected mice. Unexpectedly, neutrophils have a limited effect in controlling viral propagation upon passive immunotherapy administration, which is mostly mediated by NK cells. Instead, neutrophils operate as essential inducers of a potent host humoral antiviral response. Thus, neutrophils play an unexpected key role in protective immunity induction by antiviral mAbs. Our work opens approaches to improve antiviral immunotherapies, as it suggests that preserving neutrophil functions and counts might be required for achieving mAb-induced protective immunity.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Células Asesinas Naturales/inmunología , Virus de la Leucemia Murina , Leucemia Experimental/inmunología , Neutrófilos/inmunología , Infecciones por Retroviridae/inmunología , Infecciones Tumorales por Virus/inmunología , Replicación Viral , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Humoral , Inmunidad Innata , Inmunoterapia , Leucemia Eritroblástica Aguda/inmunología , Ratones
9.
J Immunol Methods ; 450: 90-94, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28802832

RESUMEN

Natural killer (NK) cells are capable of rapidly recognizing and efficiently killing tumor cells. This makes them a potentially promising agent for cancer immunotherapy. Additional genetic modifications of NK cells may further improve their anti-tumor efficacy. Numerous technical challenges associated with gene delivery into NK cells have significantly tempered this approach. We achieved efficient retroviral vector transduction of primary human NK cells that were stimulated by a combination of IL-2 and engineered K562 cells expressing membrane-bound IL-21. The activated NK cells were in less differentiated state and expressed NK cell activation receptors NKG2D, NKp30, CD16, and were highly HLA-DR-positive. This NK cell population was highly susceptible to the transduction by both GFP- and NGFR-expressing retroviral vectors, with transduction efficiency exceeding 50%. More mature CD57+ NK cell population was generally resistant to retroviral vector transduction because of poor response to the stimulation. Our findings may facilitate retroviral vector-mediated genetic engineering of human primary NK cells for future immunotherapies.


Asunto(s)
Vectores Genéticos , Inmunoterapia/métodos , Interleucina-2/metabolismo , Interleucinas/metabolismo , Células Asesinas Naturales/metabolismo , Leucemia Eritroblástica Aguda/terapia , Activación de Linfocitos , Retroviridae/genética , Transducción Genética , Transfección/métodos , Antígenos CD57/inmunología , Antígenos CD57/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Células Nutrientes , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-2/inmunología , Interleucinas/inmunología , Células Jurkat , Células K562 , Células Asesinas Naturales/inmunología , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptor 3 Gatillante de la Citotoxidad Natural/inmunología , Receptor 3 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
10.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(3): 678-682, 2017 Jun.
Artículo en Chino | MEDLINE | ID: mdl-28641617

RESUMEN

OBJECTIVE: To investigate the biological characteristics and therapeutic efficacyt of acute erythroleukemia (AEL,AML-M6). METHODS: Blood cell count, liver function, lactate dehydrogenase level, coagulation, morphology, immunology, cell genetics and molecular biology were retrospectively analyzed in 103 cases of acute erythroleukemia patients admitted in our department from May 2016 to June 2009. The therapeutic efficacy was observed by means of remission rate, relapse rate, relapse-free survival and overall survival. RESULTS: The medians of white blood cells, granulocyte, hemoglobin and platelet were 3.04×109/L, 0.67×109/L, 66 g/L, and 45×109/L,respectively. Nucleated red blood cells were found in the peripheral blood smears from 71.1% of AEL patients. None of the patients showed abnormal coagulation function. Flow cytometry analysis indicated that CD13 (93.5%),CD117(89.1%), HLA-DR(87.0%), and CD34 (80.0%) were highly expressed in AEL, and lymphoid antigens of CD4 (42.9%) and CD7(28.9%) were expressed in partial patients. Karyotype analysis in 82 patients showed 52.4% (43/82) normal karyotype, 41.5% (34/82) abnormal karyotype, and 6.1% (5/82) failed tests. In the 34 cases with abnormal karyotype, there were 14(41.2%) cases with simple chromosomal abnomality and 20(58.8%) cases with complex karyotype. The positive rate of fusion gene accounted for 16.7% in 60 patients, and the gene mutations accounted for 77.8% in 27 patients. Among 103 cases of AEL, 81 cases were treated with chemotherapy, but 66 cases can be used for therapeutic analysis, as a results the total complete remission rate derived from 2 courses of treatment was 45.5% (30/66). The relapse rate was 36.7% (11/30), and the median relapse time was 15.5 months (6.2-50 months). The median survival time of 66 patients for therapeutic analysis was 29 months. The median survival time of CR patients was very significantly longer than that of the non-CR patients(P=0.001). The 5 year survival rate of CR patients was 65%, the median time of relapse-free survival (RFS) was 46.2 months and 3-years RFS was 58%. CONCLUSION: AEL is characterized by the highly expressed CD34 antigen, and complex karyotype. Although AEL has lower CR rate and poor prognosis, CR patients can achieve long-term survival and have good quality of life.


Asunto(s)
Antígenos CD34/metabolismo , Leucemia Eritroblástica Aguda/terapia , Humanos , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/patología , Leucemia Mieloide Aguda , Pronóstico , Calidad de Vida , Estudios Retrospectivos
11.
Asian Pac J Cancer Prev ; 15(18): 7857-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25292077

RESUMEN

Natural killer (NK) cells play an important role in anti-tumor immunity. Interleukin (IL)-18 is an immunoregulatory cytokine that induces potent NK cell-dependent anti-tumor responses when administrated with other cytokines. In this study, we explored the effects of combining IL-18 and IL-2 on NK cytotoxicity as well as expression levels of the NK cell receptor NKG2D in vitro. Freshly isolated PBMCs were incubated for 48 h with IL-18 and IL-2, then CD107a expression on CD3-CD56+ NK cells was determined by three-colour flow cytometry to evaluate the cytotoxicity of NK cells against human erythroleukemia K562 cells and human colon carcinoma HT29 cells. Flow cytometric analysis was also employed to determine NKG2D expression on NK cells. The combined use of IL-18 and IL-2 significantly increased CD107a expression on NK cells compared with using IL-18 or IL-2 alone, suggesting that the combination of these two cytokines exerted synergistic enhancement of NK cytotoxicity. IL-18 also enhanced NKG2D expression on NK cells when administered with IL-2. In addition, blockade of NKG2D signaling with NKG2D-blocking antibody attenuated the up-regulatory effect of combining IL-18 and IL-2 on NK cytolysis. Our data revealed that IL-18 synergized with IL-2 to dramatically enhance the cytolytic activity of human NK cells in a NKG2D-dependent manner. The results appear encouraging for the use of combined IL-18 and IL-2 in tumor immunotherapy.


Asunto(s)
Neoplasias del Colon/patología , Citotoxicidad Inmunológica , Sinergismo Farmacológico , Interleucina-18/farmacología , Interleucina-2/farmacología , Células Asesinas Naturales/efectos de los fármacos , Leucemia Eritroblástica Aguda/patología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Citometría de Flujo , Humanos , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Células Tumorales Cultivadas
12.
J Leukoc Biol ; 96(3): 397-410, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24823810

RESUMEN

CD8(+) T cells must detect foreign antigens and differentiate into effector cells to eliminate infections. But, when self-antigen is recognized instead, mechanisms of peripheral tolerance prevent acquisition of effector function to avoid autoimmunity. These distinct responses are influenced by inflammatory and regulatory clues from the tissue environment, but the mechanism(s) by which naive T cells interpret these signals to generate the appropriate immune response are unclear. The identification of the molecules operative in these cell-fate decisions is crucial for developing new treatment options for patients with cancer or autoimmunity, where manipulation of T cell activity is desired to alter the course of disease. With the use of an in vivo murine model to examine CD8(+) T cell responses to healthy self-tissue, we correlated self-tolerance with a failure to induce the T-box transcription factors T-bet and Eomes. However, inflammation associated with acute microbial infection induced T-bet and Eomes expression and promoted effector differentiation of self-reactive T cells under conditions that normally favor tolerance. In the context of a Listeria infection, these functional responses relied on elevated T-bet expression, independent of Eomes. Alternatively, infection with LCMV induced higher Eomes expression, which was sufficient in the absence of T-bet to promote effector cytokine production. Our results place T-box transcription factors at a molecular crossroads between CD8(+) T cell anergy and effector function upon recognition of peripheral self-antigen, and suggest that inflammation during T cell priming directs these distinct cellular responses.


Asunto(s)
Supresión Clonal/inmunología , Inflamación/inmunología , Autotolerancia/inmunología , Proteínas de Dominio T Box/fisiología , Linfocitos T/inmunología , Animales , Autoantígenos/inmunología , Diferenciación Celular , Línea Celular Tumoral , Anergia Clonal/inmunología , Citocinas/biosíntesis , Citocinas/genética , Regulación de la Expresión Génica/inmunología , Genes RAG-1 , Inmunización , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/terapia , Listeria/inmunología , Listeriosis/inmunología , Ratones , Ratones Noqueados , Bazo/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Especificidad del Receptor de Antígeno de Linfocitos T
13.
Leukemia ; 28(11): 2213-21, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24705479

RESUMEN

Acute myeloid leukemia (AML) is a biologically heterogeneous group of related diseases in urgent need of better therapeutic options. Despite this heterogeneity, overexpression of the interleukin (IL)-3 receptor α-chain (IL-3 Rα/CD123) on both the blast and leukemic stem cell (LSC) populations is a common occurrence, a finding that has generated wide interest in devising new therapeutic approaches that target CD123 in AML patients. We report here the development of CSL362, a monoclonal antibody to CD123 that has been humanized, affinity-matured and Fc-engineered for increased affinity for human CD16 (FcγRIIIa). In vitro studies demonstrated that CSL362 potently induces antibody-dependent cell-mediated cytotoxicity of both AML blasts and CD34(+)CD38(-)CD123(+) LSC by NK cells. Importantly, CSL362 was highly effective in vivo reducing leukemic cell growth in AML xenograft mouse models and potently depleting plasmacytoid dendritic cells and basophils in cynomolgus monkeys. Significantly, we demonstrated CSL362-dependent autologous depletion of AML blasts ex vivo, indicating that CSL362 enables the efficient killing of AML cells by the patient's own NK cells. These studies offer a new therapeutic option for AML patients with adequate NK-cell function and warrant the clinical development of CSL362 for the treatment of AML.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/inmunología , Humanos , Células Asesinas Naturales/inmunología , Leucemia Eritroblástica Aguda/inmunología , Leucemia Mieloide Aguda/inmunología , Macaca fascicularis , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Ingeniería de Proteínas , Receptores de IgG/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Immunol ; 287(2): 106-11, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24487034

RESUMEN

Different molecules regulate the response of tumoral tissues to ionizing radiation. The objective of this work was to determine if HLA-G1 expression modulates the radiosensitivity of human tumoral cell lines. To this end, human melanoma M8 and human erythroleukemia K562 cell lines, with their correspondent HLA-G1 negative and positive variants, were gamma irradiated and the survival frequency was determined by clonogenic assay. The survival fraction of HLA-G1 expressing cells was around 60% of HLA-G1 negative cells. The generation of acidic vesicular organelles was higher in HLA-G1 positive cells. Apoptosis levels showed statistically significant differences only in K562 cells, whereas the variation in G2/M cycle progression was only significant in M8 cells. In addition, irradiation diminished cell-surface HLA-G1 and increased soluble HLA-G1 levels. Soluble HLA-G1 has no influence on cell survival in any cell line. In summary, we could demonstrate that HLA-G1 confers higher radiosensitivity to HLA-G1 expressing cells.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Antígenos HLA-G/metabolismo , Leucemia Eritroblástica Aguda/radioterapia , Melanoma/radioterapia , Tolerancia a Radiación , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular , Rayos gamma , Antígenos HLA-G/genética , Humanos , Células K562 , Leucemia Eritroblástica Aguda/inmunología , Melanoma/inmunología
15.
Blood ; 123(8): 1218-28, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24363400

RESUMEN

Despite the remarkable efficacy of tyrosine kinase inhibitors (TKIs) in eliminating differentiated chronic myeloid leukemia (CML) cells, recent evidence suggests that leukemic stem and progenitor cells (LSPCs) persist long term, which may be partly attributable to cytokine-mediated resistance. We evaluated the expression of the interleukin 3 (IL-3) receptor α subunit (CD123), an established marker of acute myeloid leukemia stem cells, on CML LSPCs and the potential of targeting those cells with the humanized anti-CD123 monoclonal antibody CSL362. Compared with normal donors, CD123 expression was higher in CD34(+)/CD38(-) cells of both chronic phase and blast crisis CML patients, with levels increasing upon disease progression. CSL362 effectively targeted CML LSPCs by selective antibody-dependent cell-mediated cytotoxicity (ADCC)-facilitated lysis of CD123(+) cells and reduced leukemic engraftment in mice. Importantly, not only were healthy donor allogeneic natural killer (NK) cells able to mount an effective CSL362-mediated ADCC response, but so were CML patients' autologous NK cells. In addition, CSL362 also neutralized IL-3-mediated rescue of TKI-induced cell death. Notably, combination of TKI- and CSL362-induced ADCC caused even greater reduction of CML progenitors and further augmented their preferential elimination over normal hematopoietic stem and progenitor cells. Thus, our data support the further evaluation of CSL362 therapy in CML.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Receptores de Interleucina-3/inmunología , ADP-Ribosil Ciclasa 1/metabolismo , Adulto , Anciano , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígenos CD34/metabolismo , Línea Celular , Femenino , Humanos , Subunidad alfa del Receptor de Interleucina-3/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Células Madre/inmunología , Células Madre/metabolismo , Células Madre/patología
16.
J Virol ; 87(24): 13760-74, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109240

RESUMEN

To assess the possible contribution of host immune responses to the exertion of Fv2-associated resistance to Friend virus (FV)-induced disease development, we inoculated C57BL/6 (B6) mice that lacked various subsets of lymphocytes with FV containing no lactate dehydrogenase-elevating virus. Fv2(r) B6 mice lacking CD4(+) T cells developed early polycythemia and fatal erythroleukemia, while B6 mice lacking CD8(+) T cells remained resistant. Erythroid progenitor cells infected with spleen focus-forming virus (SFFV) were eliminated, and no polycythemia was observed in B cell-deficient B6 mice, but they later developed myeloid leukemia associated with oligoclonal integration of ecotropic Friend murine leukemia virus. Additional depletion of natural killer and/or CD8(+) T cells from B cell-deficient B6 mice resulted in the expansion of SFFV proviruses and the development of polycythemia, indicating that SFFV-infected erythroid cells are not only restricted in their growth but are actively eliminated in Fv2(r) mice through cellular immune responses.


Asunto(s)
Virus de la Leucemia Murina de Friend/inmunología , Inmunidad Celular , Inmunidad Humoral , Leucemia Eritroblástica Aguda/veterinaria , Enfermedades de los Roedores/inmunología , Animales , Linfocitos B/inmunología , Progresión de la Enfermedad , Resistencia a la Enfermedad , Femenino , Virus de la Leucemia Murina de Friend/genética , Humanos , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/virología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Enfermedades de los Roedores/virología , Virus Formadores de Foco en el Bazo/genética , Virus Formadores de Foco en el Bazo/inmunología , Linfocitos T/inmunología
17.
Cancer Immunol Immunother ; 62(2): 257-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22890822

RESUMEN

The important role of tumor-specific cytotoxic CD8(+) T cells is well defined in the immune control of the tumors, but the role of effector CD4(+) T cells is poorly understood. In the current research, we have used a murine retrovirus-induced tumor cell line of C57BL/6 mouse origin, namely FBL-3 cells, as a model to study basic mechanisms of immunological control and escape during tumor formation. This study shows that tumor-specific CD4(+) T cells are able to protect against virus-induced tumor cells. We show here that there is an expansion of tumor-specific CD4(+) T cells producing cytokines and cytotoxic molecule granzyme B (GzmB) in the early phase of tumor growth. Importantly, we demonstrate that in vivo depletion of regulatory T cells (Tregs) and CD8(+) T cells in FBL-3-bearing DEREG transgenic mice augments IL-2 and GzmB production by CD4(+) T cells and increases FV-specific CD4(+) T-cell effector and cytotoxic responses leading to the complete tumor regression. Therefore, the capacity to reject tumor acquired by tumor-reactive CD4(+) T cells largely depends on the direct suppressive activity of Tregs. We suggest that a cytotoxic CD4(+) T-cell immune response may be induced to enhance resistance against oncovirus-associated tumors.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Leucemia Eritroblástica Aguda/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular Tumoral , Citocinas/biosíntesis , Citocinas/inmunología , Citotoxicidad Inmunológica/inmunología , Femenino , Virus de la Leucemia Murina de Friend/inmunología , Granzimas/biosíntesis , Granzimas/inmunología , Leucemia Eritroblástica Aguda/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Citotóxicos/virología
18.
J Immunol Methods ; 387(1-2): 191-8, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23142458

RESUMEN

Recombinant erythropoietin (rHuEPO) is used extensively to treat anaemia associated with chronic kidney disease. However, the development of neutralizing antibodies (NAbs) to rHuEPO can result in the development of antibody-mediated pure red cell aplasia (PRCA). The detection of NAb in patient sera by in vitro bioassay relies on the inhibition of a cellular response to rHuEPO. Current bioassays for rHuEPO measure proliferation in responsive cell lines such as the erythroleukaemic cell lines, UT-7 and UT-7/EPO, the latter sensitized to EPO. Using these cell lines, we show the dose-responsive induction of both PIM1 and EGR1 gene expression in UT-7 cells and of EGR1 in UT-7/EPO cells. The expression of EGR1 in UT-7/EPO cells in response to rHuEPO was comparable to the proliferative response measured by (3)H-thymidine incorporation and could be inhibited by serum from a patient with NAb-mediated PRCA in a dilution-dependent manner. Bioassays based on the induction of endogenous gene expression are comparable to current bioassays but are considerably quicker given that incubation time is decreased from 2-3 days to 50 min. Measurement of EGR1 gene expression in response to rHuEPO in UT-7/EPO cells offers a rapid, non-radioactive and automatable alternative to current assays for the detection of rHuEPO NAbs.


Asunto(s)
Anticuerpos Neutralizantes/análisis , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Eritropoyetina/farmacología , Expresión Génica/efectos de los fármacos , Proteínas Recombinantes/farmacología , Animales , Anticuerpos Neutralizantes/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritropoyetina/genética , Eritropoyetina/inmunología , Expresión Génica/inmunología , Humanos , Sueros Inmunes/inmunología , Sueros Inmunes/farmacología , Técnicas Inmunológicas/métodos , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/patología , Aplasia Pura de Células Rojas/sangre , Aplasia Pura de Células Rojas/inmunología , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Suero/inmunología , Ovinos , Factores de Tiempo
19.
Hum Immunol ; 74(3): 286-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23238216

RESUMEN

The suppressive functions of HLA-G to various immune cells have been well established. The proportion of HLA-G expression in malignant lesion cells was found from negative to 100%. However, effects for the different proportion of HLA-G expression on the cytolysis of NK cells remain to be explored. In this study, NK cytolysis to the various proportion of HLA-G1 expression on leukemia cell line K562 was investigated. Analysis of NK cell cytotoxicity was by detecting the NK cell surface CD107a expression. Data showed that NK cell cytolysis could be inhibited by the HLA-G1 expression and in a manner of HLA-G1 expression proportion dependent manner (r = 0.925, p = 0.008). Our study provided further understanding for the roles of HLA-G1 expression in malignant cell immune escaping from NK cells.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Antígenos HLA-G/inmunología , Células Asesinas Naturales/inmunología , Leucemia Eritroblástica Aguda/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Citometría de Flujo , Antígenos HLA-G/genética , Antígenos HLA-G/metabolismo , Humanos , Células K562 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Transfección
20.
Cancer Immunol Immunother ; 61(9): 1451-61, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22310931

RESUMEN

Natural killer (NK) cells hold promise for adoptive cancer immunotherapy but are dependent on cytokines such as interleukin (IL)-2 for growth and cytotoxicity. Here, we investigated the consequences of ectopic expression of IL-15 in human NK cells. IL-2 and IL-15 belong to the common γ chain family of cytokines and have overlapping activities. Transduction of clinically applicable NK-92 cells with lentiviral vectors encoding human IL-15 resulted in predominantly intracellular expression of the cytokine, and STAT5 activation, proliferation and cytotoxicity of the producer cells in the absence of IL-2. Growth of non-transduced bystander cells was not supported, allowing rapid enrichment of gene-modified cells solely by IL-2 withdrawal. This was also the case upon transduction of NK-92 and NKL cells with a bicistronic lentiviral vector encoding IL-15 and a chimeric antigen receptor (CAR) targeting the pancarcinoma antigen EpCAM. Effector cells co-expressing CAR and IL-15 continued to proliferate in the absence of exogenous cytokines and displayed high and selective cell-killing activity against EpCAM-expressing breast carcinoma cells that were resistant to the natural cytotoxicity of unmodified NK cells. This strategy facilitates rapid isolation and continuous expansion of retargeted NK cells and may extend their potential clinical utility.


Asunto(s)
Citocinas/biosíntesis , Inmunoterapia/métodos , Interleucina-15/biosíntesis , Células Asesinas Naturales/inmunología , Receptores de Antígenos/inmunología , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Línea Celular Tumoral , Citocinas/genética , Citocinas/inmunología , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , ADN Complementario/genética , Molécula de Adhesión Celular Epitelial , Humanos , Interleucina-15/genética , Interleucina-15/inmunología , Interleucina-2/inmunología , Interleucina-2/farmacología , Células K562 , Células Asesinas Naturales/metabolismo , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/terapia , Melanoma/inmunología , Melanoma/terapia , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Antígenos/biosíntesis , Receptores de Antígenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...