Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 8(12): eabh4050, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35319989

RESUMO

Radiotherapy is a mainstay cancer therapy whose antitumor effects partially depend on T cell responses. However, the role of Natural Killer (NK) cells in radiotherapy remains unclear. Here, using a reverse translational approach, we show a central role of NK cells in the radiation-induced immune response involving a CXCL8/IL-8-dependent mechanism. In a randomized controlled pancreatic cancer trial, CXCL8 increased under radiotherapy, and NK cell positively correlated with prolonged overall survival. Accordingly, NK cells preferentially infiltrated irradiated pancreatic tumors and exhibited CD56dim-like cytotoxic transcriptomic states. In experimental models, NF-κB and mTOR orchestrated radiation-induced CXCL8 secretion from tumor cells with senescence features causing directional migration of CD56dim NK cells, thus linking senescence-associated CXCL8 release to innate immune surveillance of human tumors. Moreover, combined high-dose radiotherapy and adoptive NK cell transfer improved tumor control over monotherapies in xenografted mice, suggesting NK cells combined with radiotherapy as a rational cancer treatment strategy.


Assuntos
Interleucina-8 , Células Matadoras Naturais , Neoplasias , Transferência Adotiva , Animais , Humanos , Imunidade , Interleucina-8/imunologia , Interleucina-8/metabolismo , Células Matadoras Naturais/imunologia , Camundongos , Neoplasias/imunologia , Neoplasias/radioterapia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Eur J Immunol ; 48(2): 355-365, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29105756

RESUMO

To exploit autologous NK cells for cancer immunotherapy, it is highly relevant to circumvent killer cell immunoglobulin-like receptor (KIR)-mediated self-inhibition of human NK cells by HLA-I-expressing tumor cells. Here, we show that stimulation of NK cells with IL-12/15/18 for two days led to downregulation of surface expression of the inhibitory KIR2DL2/L3, KIR2DL1 and KIR3DL1 receptors on peripheral blood NK cells. Downregulation of KIR expression was attributed to decreased KIR mRNA levels which could be re-induced already 3 days after re-culture in IL-2. Reduced KIR2DL2/L3 expression on IL-12/15/18-activated NK cells resulted in less inhibition upon antibody-mediated KIR engagement and increased CD16-dependent cytotoxicity in redirected lysis assays. Most importantly, downregulated KIR2DL2/L3 expression enabled enhanced cytotoxicity of IL-12/15/18-stimulated NK cells against tumor cells expressing cognate HLA-I molecules. NK cells pre-activated with IL-12/15/18 were previously shown to exert potent anti-tumor activity and memory-like long-lived functionality, mediating remission in a subset of acute myeloid leukemia (AML) patients in a clinical trial. Our study reveals a novel mechanism of IL-12/15/18 in improving the cytotoxicity of NK cells by reducing their sensitivity to inhibition by self-HLA-I due to decreased KIR expression, highlighting the potency of IL-12/15/18-activated NK cells for anti-tumor immunotherapy protocols.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/terapia , Receptores KIR2DL2/metabolismo , Receptores KIR2DL3/metabolismo , Receptores KIR3DL1/metabolismo , Animais , Citotoxicidade Celular Dependente de Anticorpos , Linhagem Celular Tumoral , Regulação para Baixo , Antígenos HLA/metabolismo , Humanos , Interleucina-12/metabolismo , Interleucina-15/metabolismo , Interleucina-18/metabolismo , Células Matadoras Naturais/transplante , Leucemia Mieloide Aguda/imunologia , Ativação Linfocitária , Camundongos
3.
Cell Death Dis ; 8(8): e2973, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28771222

RESUMO

Aggressive breast cancer is associated with poor patient outcome and characterized by the development of tumor cell variants that are able to escape from control of the immune system or are resistant to targeted therapies. The complex molecular mechanisms leading to immune escape and therapy resistance are incompletely understood. We have previously shown that high miR-519a-3p levels are associated with poor survival in breast cancer. Here, we demonstrate that miR-519a-3p confers resistance to apoptosis induced by TRAIL, FasL and granzyme B/perforin by interfering with apoptosis signaling in breast cancer cells. MiR-519a-3p diminished the expression of its direct target genes for TRAIL-R2 (TNFRSF10B) and for caspase-8 (CASP8) and its indirect target gene for caspase-7 (CASP7), resulting in reduced sensitivity and tumor cell apoptosis in response to apoptotic stimuli. Furthermore, miR-519a-3p impaired tumor cell killing by natural killer (NK) cells via downregulation of the NKG2D ligands ULBP2 and MICA on the surface of tumor cells that are crucial for the recognition of these tumor cells by NK cells. We determined that miR-519a-3p was overexpressed in more aggressive mutant TP53 breast cancer that was associated with poor survival. Furthermore, low levels of TRAIL-R2, caspase-7 and caspase-8 correlated with poor survival, suggesting that the inhibitory effect of miR-519a-3p on TRAIL-R2 and caspases may have direct clinical relevance in lowering patient's prognosis. In conclusion, we demonstrate that miR-519a-3p is a critical factor in mediating resistance toward cancer cell apoptosis and impairing tumor cell recognition by NK cells. This joint regulation of apoptosis and immune cell recognition through miR-519a-3p supports the hypothesis that miRNAs are key regulators of cancer cell fate, facilitating cancer progression and evasion from immunosurveillance at multiple and interconnected levels.


Assuntos
Apoptose/imunologia , Neoplasias da Mama/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , MicroRNAs/imunologia , RNA Neoplásico/imunologia , Evasão Tumoral , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Células Matadoras Naturais/patologia , Células MCF-7 , MicroRNAs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , RNA Neoplásico/genética
4.
Nature ; 547(7662): 222-226, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28678784

RESUMO

T cells directed against mutant neo-epitopes drive cancer immunity. However, spontaneous immune recognition of mutations is inefficient. We recently introduced the concept of individualized mutanome vaccines and implemented an RNA-based poly-neo-epitope approach to mobilize immunity against a spectrum of cancer mutations. Here we report the first-in-human application of this concept in melanoma. We set up a process comprising comprehensive identification of individual mutations, computational prediction of neo-epitopes, and design and manufacturing of a vaccine unique for each patient. All patients developed T cell responses against multiple vaccine neo-epitopes at up to high single-digit percentages. Vaccine-induced T cell infiltration and neo-epitope-specific killing of autologous tumour cells were shown in post-vaccination resected metastases from two patients. The cumulative rate of metastatic events was highly significantly reduced after the start of vaccination, resulting in a sustained progression-free survival. Two of the five patients with metastatic disease experienced vaccine-related objective responses. One of these patients had a late relapse owing to outgrowth of ß2-microglobulin-deficient melanoma cells as an acquired resistance mechanism. A third patient developed a complete response to vaccination in combination with PD-1 blockade therapy. Our study demonstrates that individual mutations can be exploited, thereby opening a path to personalized immunotherapy for patients with cancer.


Assuntos
Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Melanoma/imunologia , Melanoma/terapia , Mutação/genética , Medicina de Precisão/métodos , RNA/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antígeno B7-H1/imunologia , Antígenos CD8/imunologia , Vacinas Anticâncer/uso terapêutico , Epitopos/genética , Epitopos/imunologia , Humanos , Imunoterapia/métodos , Melanoma/genética , Metástase Neoplásica , Recidiva Local de Neoplasia/prevenção & controle , Nivolumabe , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Linfócitos T/imunologia , Vacinação , Microglobulina beta-2/deficiência
6.
Oncoimmunology ; 5(9): e1219007, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27757317

RESUMO

Natural killer (NK) cells are promising antitumor effector cells, but the generation of sufficient NK cell numbers for adoptive immunotherapy remains challenging. Therefore, we developed a method for highly efficient ex vivo expansion of human NK cells. Ex vivo expansion of NK cells in medium containing IL-2 and irradiated clinical-grade feeder cells (EBV-LCL) induced a 22-fold NK cell expansion after one week that was significantly increased to 53-fold by IL-21. Repeated stimulation with irradiated EBV-LCL and IL-2 and addition of IL-21 at the initiation of the culture allowed sustained NK cell proliferation with 1011-fold NK cell expansion after 6 weeks. Compared to naive NK cells, expanded NK cells upregulated TRAIL, NKG2D, and DNAM-1, had superior cytotoxicity against tumor cell lines in vitro and produced more IFNγ and TNF-α upon PMA/Iono stimulation. Most importantly, adoptive transfer of NK cells expanded using feeder cells, IL-2 and IL-21 led to significant inhibition of tumor growth in a melanoma xenograft mouse model, which was greater than with NK cells activated with IL-2 alone. Intriguingly, adoptively transferred NK cells maintained their enhanced production of IFNγ and TNF-α upon ex vivo restimulation, although they rapidly lost their capacity to degranulate and mediate tumor cytotoxicity after the in vivo transfer. In conclusion, we developed a protocol for ex vivo NK cell expansion that results in outstanding cell yields. The expanded NK cells possess potent antitumor activity in vitro and in vivo and could be utilized at high numbers for adoptive immunotherapy in the clinic.

7.
Oncoimmunology ; 5(9): e1219009, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27757318

RESUMO

Natural killer (NK) cell infusions can induce remissions in subsets of patients with different types of cancer. The optimal strategies for NK cell activation prior to infusion are still under debate. There is recent evidence that NK cells can acquire long-term functional competence by preactivation with the cytokines IL-12/15/18. The mechanisms supporting the maintenance of long-term NK cell antitumor activity are incompletely under-stood. Here, we show that NK cells preactivated in vitro with IL-12/15/18, but not with IL-15 alone, maintained high antitumor activity even 1 mo after transfer into lymphopenic RAG-2-/-γc-/- mice. The NK cell intrinsic ability for IFNγ production coincided with demethylation of the conserved non-coding sequence (CNS) 1 in the Ifng locus, previously shown to enhance transcription of Ifng. In a xenograft melanoma mouse model, human IL-12/15/18-preactivated NK cells rejected tumors more efficiently. In RAG-2-/-γc-/- mice, co-transfer of CD4+ T cells further improved the long-term competence of NK cells for IFNγ production that was dependent on IL-2. CD4+ T cell activation during homeostatic proliferation required macrophages and further promoted the long-term NK cell antitumor activity. Thus, NK cells can "remember" a previous exposure to cytokines by epigenetic imprinting resulting in a remarkable stability of the IFNγ-producing phenotype after adoptive transfer. In addition, our results support combination of cytokine-preactivated NK cells with CD4+ T cell activation upon lymphopenic conditioning to achieve long-term NK cell effector function for cancer immunotherapy.

9.
Nat Commun ; 7: 10764, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26948869

RESUMO

The high-mobility group box 1 (HMGB1) protein has a central role in immunological antitumour defense. Here we show that natural killer cell-derived HMGB1 directly eliminates cancer cells by triggering metabolic cell death. HMGB1 allosterically inhibits the tetrameric pyruvate kinase isoform M2, thus blocking glucose-driven aerobic respiration. This results in a rapid metabolic shift forcing cells to rely solely on glycolysis for the maintenance of energy production. Cancer cells can acquire resistance to HMGB1 by increasing glycolysis using the dimeric form of PKM2, and employing glutaminolysis. Consistently, we observe an increase in the expression of a key enzyme of glutaminolysis, malic enzyme 1, in advanced colon cancer. Moreover, pharmaceutical inhibition of glutaminolysis sensitizes tumour cells to HMGB1 providing a basis for a therapeutic strategy for treating cancer.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/fisiopatologia , Proteína HMGB1/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Morte Celular , Linhagem Celular Tumoral , Respiração Celular , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Glucose/metabolismo , Glicólise , Proteína HMGB1/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
10.
J Dtsch Dermatol Ges ; 13(1): 23-9, 2015 Jan.
Artigo em Inglês, Alemão | MEDLINE | ID: mdl-25640488

RESUMO

During the recent years, immunotherapy has obtained substantial impact on the clinical treatment of melanoma. Besides promising approaches based on T lymphocytes, natural killer (NK) cells have gained more and more attention as anti-melanoma effector cells. NK cell activation is inhibited by HLA class I molecules expressed by target cells, so they preferentially attack tumor cells that express low levels of HLA class I. Partial or complete loss of HLA class I expression is a frequent event during the development of melanoma. In parallel, ligands for activating NK cell receptors become induced upon malignant transformation. Thus, melanoma cells are often efficiently recognized and lysed by NK cells at least in vitro. In vivo, however, melanomas have developed multiple sophisticated strategies to escape from NK cell mediated attack. Several novel approaches aim at harnessing NK cells to treat melanoma patients and to counteract existing tumor escape mechanisms. This review summarizes the most recent advances in the field.


Assuntos
Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Melanoma/imunologia , Melanoma/terapia , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Humanos , Modelos Imunológicos
11.
Oncoimmunology ; 2(4): e23811, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23734329

RESUMO

The adoptive transfer of interleukin (IL)-2-expanded natural killer (NK) cells has provided unsatisfactory clinical benefits to patients affected by solid tumors. Our study demonstrates that the activation of NK cells with IL-12/IL-15/IL-18 prior to transfer into tumor-bearing mice is critical for obtaining high recovery rates, effector functions in vivo and tumor regression.

12.
J Exp Med ; 209(13): 2351-65, 2012 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-23209317

RESUMO

Natural killer cell (NK cell)-based immunotherapy of cancer is hampered by the transient effector function of NK cells. Recently, mouse IL-12/15/18-preactivated NK cells were shown to persist with sustained effector function in vivo. Our study investigated the antitumor activity of such NK cells. A single injection of syngeneic IL-12/15/18-preactivated NK cells, but neither naive nor IL-15- or IL-2-pretreated NK cells, combined with irradiation substantially reduced growth of established mouse tumors. Radiation therapy (RT) was essential for the antitumor activity of transferred NK cells. IL-12/15/18-preactivated NK cells expressed high levels of IL-2Rα (CD25), and their rapid in vivo proliferation depended on IL-2 produced by CD4+ T cells. IL-12/15/18-preactivated NK cells accumulated in the tumor tissue and persisted at high cell numbers with potent effector function that required the presence of CD4+ T cells. RT greatly increased numbers and function of transferred NK cells. Human IL-12/15/18-preactivated NK cells also displayed sustained effector function in vitro. Our study provides a better understanding for the rational design of immunotherapies of cancer that incorporate NK cells. Moreover, our results reveal an essential role of CD4+ T cell help for sustained antitumor activity by NK cells linking adaptive and innate immunity.


Assuntos
Imunoterapia Adotiva , Células Matadoras Naturais/imunologia , Neoplasias Experimentais/terapia , Imunidade Adaptativa , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada , Humanos , Imunidade Inata , Interferon gama/biossíntese , Interferon gama/deficiência , Interferon gama/genética , Interleucina-12/administração & dosagem , Interleucina-15/administração & dosagem , Interleucina-18/administração & dosagem , Interleucina-2/deficiência , Interleucina-2/genética , Interleucina-2/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/patologia , Linfoma/imunologia , Linfoma/radioterapia , Linfoma/terapia , Melanoma Experimental/imunologia , Melanoma Experimental/radioterapia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/radioterapia , Baço/imunologia , Fatores de Tempo
13.
Curr Pharm Des ; 15(28): 3221-36, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19860672

RESUMO

Short peptides derived from cellular proteins may escape complete destruction during protein catabolism and finally serve as a showcase in the immune system. Exposed at the cell surface to scrutiny by T cells, MHC:peptide complexes mediate a highly specific and immediate information transfer from diseased cells to the cellular immune system. Numerous clinical vaccination trials have been carried out employing MHC-presented peptides for T-cell activation with encouraging results but so far without a final breakthrough. In this review, we briefly highlight the molecular basis of MHC-peptide interactions governed by specificity pockets and anchor residues, as summarized in allele-specific peptide motifs. State-of-the-art technology is comprehensively presented and gives an overview of modern mass spectrometric strategies used for qualitative and quantitative analysis of MHC ligands. We describe the details of the HLA-B*3801 peptide motif by comparing features of natural MHC ligands, resulting in a scoring matrix that enables epitope prediction from any viral or tumor antigen. The pronounced individuality in peptide presentation by MHC molecules, as reflected in the highly specific peptide motifs of different MHC allotypes or the tissue-specific MHC ligandomes, represents a current area of interest within this field. Finally, the identification of post-translational modifications--most important phosphorylations--and the promises this holds will be discussed in this chapter.


Assuntos
Vacinas Anticâncer/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Animais , Desenho de Fármacos , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Genes MHC Classe I/genética , Genes MHC Classe I/fisiologia , Genes MHC da Classe II/genética , Genes MHC da Classe II/fisiologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Ligantes , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA