Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(6): 106864, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37255666

RESUMO

Diffuse large B cell lymphoma (DLBCL) is one of the most common types of aggressive lymphoid malignancies. Here, we explore the contribution of RNA editing to DLBCL pathogenesis. We observed that DNA mutations and RNA editing events are often mutually exclusive, suggesting that tumors can modulate pathway outcomes by altering sequences at either the genomic or the transcriptomic level. RNA editing targets transcripts within known disease-driving pathways such as apoptosis, p53 and NF-κB signaling, as well as the RIG-I-like pathway. In this context, we show that ADAR1-mediated editing within MAVS transcript positively correlates with MAVS protein expression levels, associating with increased interferon/NF-κB signaling and T cell exhaustion. Finally, using targeted RNA base editing tools to restore editing within MAVS 3'UTR in ADAR1-deficient cells, we demonstrate that editing is likely to be causal to an increase in downstream signaling in the absence of activation by canonical nucleic acid receptor sensing.

2.
Front Cell Dev Biol ; 10: 1080626, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36684421

RESUMO

Despite hundreds of RNA modifications described to date, only RNA editing results in a change in the nucleotide sequence of RNA molecules compared to the genome. In mammals, two kinds of RNA editing have been described so far, adenosine to inosine (A-to-I) and cytidine to uridine (C-to-U) editing. Recent improvements in RNA sequencing technologies have led to the discovery of a continuously growing number of editing sites. These methods are powerful but not error-free, making routine validation of newly-described editing sites necessary. During one of these validations on DDX58 mRNA, along with A-to-I RNA editing sites, we encountered putative U-to-C editing. These U-to-C edits were present in several cell lines and appeared regulated in response to specific environmental stimuli. The same findings were also observed for the human long intergenic non-coding RNA p21 (hLincRNA-p21). A more in-depth analysis revealed that putative U-to-C edits result from A-to-I editing on overlapping antisense RNAs that are transcribed from the same loci. Such editing events, occurring on overlapping genes transcribed in opposite directions, have recently been demonstrated to be immunogenic and have been linked with autoimmune and immune-related diseases. Our findings, also confirmed by deep transcriptome data, demonstrate that such loci can be recognized simply through the presence of A-to-I and U-to-C mismatches within the same locus, reflective A-to-I editing both in the sense-oriented transcript and in the cis-natural antisense transcript (cis-NAT), implying that such clusters could be a mark of functionally relevant ADAR1 editing events.

3.
Mol Ther Nucleic Acids ; 25: 515-523, 2021 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-34589274

RESUMO

We present MultiEditR (Multiple Edit Deconvolution by Inference of Traces in R), the first algorithm specifically designed to detect and quantify RNA editing from Sanger sequencing (z.umn.edu/multieditr). Although RNA editing is routinely evaluated by measuring the heights of peaks from Sanger sequencing traces, the accuracy and precision of this approach has yet to be evaluated against gold standard next-generation sequencing methods. Through a comprehensive comparison to RNA sequencing (RNA-seq) and amplicon-based deep sequencing, we show that MultiEditR is accurate, precise, and reliable for detecting endogenous and programmable RNA editing.

4.
Immunity ; 52(6): 1075-1087.e8, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32445619

RESUMO

Enhancing immune cell functions in tumors remains a major challenge in cancer immunotherapy. Hypoxia is a common feature of solid tumors, and cells adapt by upregulating the transcription factor HIF-1α. Here, we defined the transcriptional landscape of mouse tumor-infiltrating natural killer (NK) cells by using single-cell RNA sequencing. Conditional deletion of Hif1a in NK cells resulted in reduced tumor growth, elevated expression of activation markers, effector molecules, and an enriched NF-κB pathway in tumor-infiltrating NK cells. Interleukin-18 (IL-18) from myeloid cells was required for NF-κB activation and the enhanced anti-tumor activity of Hif1a-/- NK cells. Extended culture with an HIF-1α inhibitor increased human NK cell responses. Low HIF1A expression was associated with high expression of IFNG in human tumor-infiltrating NK cells, and an enriched NK-IL18-IFNG signature in solid tumors correlated with increased overall patient survival. Thus, inhibition of HIF-1α unleashes NK cell anti-tumor activity and could be exploited for cancer therapy.


Assuntos
Citotoxicidade Imunológica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Animais , Biomarcadores , Biologia Computacional , Citocinas/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/mortalidade , Prognóstico , Análise de Célula Única , Transcriptoma , Microambiente Tumoral/imunologia
5.
Cancer Immunol Res ; 6(5): 517-527, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29514797

RESUMO

CD16A is a potent cytotoxicity receptor on human natural killer (NK) cells, which can be exploited by therapeutic bispecific antibodies. So far, the effects of CD16A-mediated activation on NK cell effector functions beyond classical antibody-dependent cytotoxicity have remained poorly elucidated. Here, we investigated NK cell responses after exposure to therapeutic antibodies such as the tetravalent bispecific antibody AFM13 (CD30/CD16A), designed for the treatment of Hodgkin lymphoma and other CD30+ lymphomas. Our results reveal that CD16A engagement enhanced subsequent IL2- and IL15-driven NK cell proliferation and expansion. This effect involved the upregulation of CD25 (IL2Rα) and CD132 (γc) on NK cells, resulting in increased sensitivity to low-dose IL2 or to IL15. CD16A engagement initially induced NK cell cytotoxicity. The lower NK cell reactivity observed 1 day after CD16A engagement could be recovered by reculture in IL2 or IL15. After reculture in IL2 or IL15, these CD16A-experienced NK cells exerted more vigorous IFNγ production upon restimulation with tumor cells or cytokines. Importantly, after reculture, CD16A-experienced NK cells also exerted increased cytotoxicity toward different tumor targets, mainly through the activating NK cell receptor NKG2D. Our findings uncover a role for CD16A engagement in priming NK cell responses to restimulation by cytokines and tumor cells, indicative of a memory-like functionality. Our study suggests that combination of AFM13 with IL2 or IL15 may boost NK cell antitumor activity in patients by expanding tumor-reactive NK cells and enhancing NK cell reactivity, even upon repeated tumor encounters. Cancer Immunol Res; 6(5); 517-27. ©2018 AACR.


Assuntos
Proliferação de Células , Citotoxicidade Imunológica/fisiologia , Memória Imunológica/fisiologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/fisiologia , Neoplasias/imunologia , Receptores de IgG/imunologia , Adulto , Animais , Anticorpos Biespecíficos/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Memória Imunológica/efeitos dos fármacos , Imunoterapia/métodos , Células Jurkat , Células K562 , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Neoplasias/terapia , Receptores de IgG/metabolismo
6.
Oncoimmunology ; 5(7): e1116674, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27622013

RESUMO

Natural Killer (NK) cells are innate effector cells that are able to recognize and eliminate tumor cells through engagement of their surface receptors. NKp30 is a potent activating NK cell receptor that elicits efficient NK cell-mediated target cell killing. Recently, B7-H6 was identified as tumor cell surface expressed ligand for NKp30. Enhanced B7-H6 mRNA levels are frequently detected in tumor compared to healthy tissues. To gain insight in the regulation of expression of B7-H6 in tumors, we investigated transcriptional mechanisms driving B7-H6 expression by promoter analyses. Using luciferase reporter assays and chromatin immunoprecipitation we mapped a functional binding site for Myc, a proto-oncogene overexpressed in certain tumors, in the B7-H6 promoter. Pharmacological inhibition or siRNA/shRNA-mediated knock-down of c-Myc or N-Myc significantly decreased B7-H6 expression on a variety of tumor cells including melanoma, pancreatic carcinoma and neuroblastoma cell lines. In tumor cell lines from different origin and primary tumor tissues of hepatocellular carcinoma (HCC), lymphoma and neuroblastoma, mRNA levels of c-Myc positively correlated with B7-H6 expression. Most importantly, upon inhibition or knock-down of c-Myc in tumor cells impaired NKp30-mediated degranulation of NK cells was observed. Thus, our data imply that Myc driven tumors could be targets for cancer immunotherapy exploiting the NKp30/B7-H6 axis.

7.
Cancer Res ; 74(13): 3429-40, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24780758

RESUMO

Natural killer (NK) cells are potent immune effector cells capable of mediating antitumor responses. Thus, during immunoediting, tumor cell populations evolve strategies to escape NK-cell-mediated recognition. In this study, we report a novel mechanism of immune escape involving tumor cell shedding of B7-H6, a ligand for the activating receptor NKp30 that mediates NK-cell binding and NK-cell-mediated killing. Tumor cells from different cancer entities released B7-H6 by ectodomain shedding mediated by the cell surface proteases "a disintegrin and metalloproteases" (ADAM)-10 and ADAM-17, as demonstrated through the use of pharmacologic inhibitors or siRNA-mediated gene attenuation. Inhibiting this proteolytic shedding process increased the levels of B7-H6 expressed on the surface of tumor cells, enhancing NKp30-mediated activation of NK cells. Notably, we documented elevated levels of soluble B7-H6 levels in blood sera obtained from a subset of patients with malignant melanoma, compared with healthy control individuals, along with evidence of elevated B7-H6 expression in melanoma specimens in situ. Taken together, our results illustrated a novel mechanism of immune escape in which tumor cells impede NK-mediated recognition by metalloprotease-mediated shedding of B7-H6. One implication of our findings is that therapeutic inhibition of specific metalloproteases may help support NK-cell-based cancer therapy.


Assuntos
Proteínas ADAM/biossíntese , Secretases da Proteína Precursora do Amiloide/biossíntese , Antígenos B7 , Células Matadoras Naturais/imunologia , Proteínas de Membrana/biossíntese , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Proteína ADAM10 , Proteína ADAM17 , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Antígenos B7/sangue , Antígenos B7/genética , Antígenos B7/metabolismo , Linhagem Celular Tumoral , Dipeptídeos/farmacologia , Células HCT116 , Células HeLa , Humanos , Ácidos Hidroxâmicos/farmacologia , Ativação Linfocitária/imunologia , Células MCF-7 , Melanoma/sangue , Melanoma/patologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Receptor 3 Desencadeador da Citotoxicidade Natural/imunologia , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Inibidores de Proteases/farmacologia , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Tiofenos/farmacologia , Evasão Tumoral , Regulação para Cima/efeitos dos fármacos
8.
Blood ; 122(5): 684-93, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23801635

RESUMO

Natural killer (NK) cells are central effector cells during innate immune responses against cancer. Natural cytotoxicity receptors expressed by NK cells such as NKp30 are involved in the recognition of transformed cells. Recently, the novel B7 family member B7-H6, which is expressed on the cell surface of various tumor cells including hematological malignancies, was identified as an activating ligand for NKp30. To investigate expression and regulation of B7-H6, we generated monoclonal antibodies. Our study reveals that B7-H6 surface protein and messenger RNA (mRNA) expression in various tumor cell lines was downregulated upon treatment with pan- or class I histone deacetylase inhibitors (HDACi) as well as after small interfering RNA-mediated knockdown of the class I histone deacetylases (HDAC) 2 or 3. B7-H6 downregulation was associated with decreased B7-H6 reporter activity and reduced histone acetylation at the B7-H6 promoter. In certain primary lymphoma and hepatocellular carcinoma samples, B7-H6 mRNA levels were elevated and correlated with HDAC3 expression. Finally, downregulation of B7-H6 on tumor cells by HDACi reduced NKp30-dependent effector functions of NK cells. Thus, we identified a novel mechanism that governs B7-H6 expression in tumor cells that has implications for potential cancer treatments combining immunotherapy with HDACi.


Assuntos
Antígenos B7/genética , Inibidores de Histona Desacetilases/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Receptor 3 Desencadeador da Citotoxicidade Natural/metabolismo , Neoplasias/imunologia , Antígenos B7/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Células Hep G2 , Humanos , Células K562 , Células Matadoras Naturais/imunologia , Ligantes , Receptor 3 Desencadeador da Citotoxicidade Natural/agonistas , Neoplasias/genética , Neoplasias/metabolismo , Transfecção , Células Tumorais Cultivadas , Evasão Tumoral/efeitos dos fármacos , Evasão Tumoral/genética , Evasão Tumoral/imunologia
9.
Int J Oncol ; 40(3): 840-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22102168

RESUMO

Newcastle disease virus (NDV) is a negative sense RNA paramyxovirus of birds which in human tumor cells, in contrast to human non-tumor cells, has shown replication competence leading to tumor cell death (i.e., tumor selectivity and viral oncolysis). Our study demonstrates that this virus induces high levels of pro-inflammatory cytokines in the bronchial lavage fluid of mice after nasal application and also in vitro in human dendritic cells (DCs). NDV is known as a very efficient inductor of type I interferon (IFN). The presented data show the key role played by the cell surface receptor to type I IFN (IFNAR) but not by the interferon transcription factors IRF-3 and IRF-7 in the induction of the important pro-inflammatory cytokine IL-12 upon transcription of NDV genes in DCs. We show that NDV activates in infected cells the helicase RIG-I. In Tregs, the activation of RIG-I was shown in other studies to inhibit the suppressive function of these cells. We thus conclude that NDV in tumor therapy may help to stimulate T effector cells but also to block Treg cells, thereby alleviating a brake to antitumor activity.


Assuntos
Células Dendríticas/virologia , Inflamação/virologia , Interferon-alfa/imunologia , Vírus da Doença de Newcastle/imunologia , Vírus Oncolíticos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/virologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , RNA Helicases DEAD-box/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/imunologia , Interferon-alfa/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Receptores Imunológicos , Replicação Viral/imunologia
10.
Cancer Res ; 71(18): 5998-6009, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21764762

RESUMO

Natural killer (NK) cells are immune cells sensing and eliminating foreign, stressed, transformed, and senescent cells through specialized surface receptors, such as NKG2D, that interacts with several virus- or stress-inducible ligands, including ULBP1 and -2, which are expressed on target cell surfaces. For example, induction of DNA damage or cellular senescence pathways in tumor cells led to upregulation of NKG2D ligands that activate NK cells. Although, both pathways activate p53, the relationship of p53 activation to upregulation of NKG2D ligands has not been addressed. In this study, we report that induction of wild-type p53, but not mutant p53, strongly upregulated mRNA and cell surface expression of ULBP1 and -2, whereas expression of other NK cell ligands was not affected. We defined intronic p53-responsive elements in these two novel p53 target genes. Coculture of wild-type p53-induced human tumor cells with primary human NK cells enhanced NKG2D-dependent degranulation and IFN-γ production by NK cells. Accordingly, treatment of certain wild-type p53-expressing tumor cell lines with the p53-reactivating small molecular compound RITA resulted in upregulation of ULBP2 mRNA and cell surface protein expression. Taken together, our findings define the involvement of p53 in the regulation of specific NKG2D ligands that enhance NK cell-mediated target recognition. One implication of our work is that activating p53 after adoptive transfer of NK cells might constitute an effective combinatorial strategy of NK cell-based immunochemotherapy in cancers in which wild-type p53 function is preserved.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Células Matadoras Naturais/imunologia , Neoplasias Pulmonares/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Linhagem Celular Tumoral , Furanos/farmacologia , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Íntrons , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/imunologia , Regulação para Cima/efeitos dos fármacos
11.
Vaccine ; 27(40): 5480-7, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19616491

RESUMO

We describe for the first time a short sequence of the CD11c promoter (700 bp, named as CD11cS) which induces selective antigen expression in dendritic cells (DCs). It showed a stronger promoter activity than the hitherto used long CD11c promoter (5.5 kb, named as CD11cL), which had been reported inefficient to induce immune responses. After application to the ear pinna and electroporation (EP), CD11cS based DNA vaccines induced specific B- and T-cell responses, including IFN-gamma secretion. Such vaccines achieved induction of anti-tumor immunity comparable in strength to vaccines having the strong tissue non-specific CMV promoter, in both prophylactic and therapeutic settings of mouse tumor models. This short CD11c promoter appears to be a safe and a practical tool for DC-specific gene targeting and for vaccination.


Assuntos
Antígeno CD11c/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Regiões Promotoras Genéticas , Vacinas de DNA/imunologia , Animais , Linfócitos B/imunologia , Antígeno CD11c/genética , Linhagem Celular Tumoral , Feminino , Interferon gama/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias Experimentais/terapia , Linfócitos T/imunologia , Transfecção
12.
J Virol ; 83(16): 8108-21, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19515783

RESUMO

The avian paramyxovirus Newcastle disease virus (NDV) selectively replicates in tumor cells and is known to stimulate T-cell-, macrophage-, and NK cell-mediated responses. The mechanisms of NK cell activation by NDV are poorly understood so far. We studied the expression of ligand structures for activating NK cell receptors on NDV-infected tumor cells. Upon infection with the nonlytic NDV strain Ulster and the lytic strain MTH-68/H, human carcinoma and melanoma cells showed enhanced expression of ligands for the natural cytotoxicity receptors NKp44 and NKp46, but not NKp30. Ligands for the activating receptor NKG2D were partially downregulated. Soluble NKp44-Fc and NKp46-Fc, but not NKp30-Fc, chimeric proteins bound specifically to NDV-infected tumor cells and to NDV particle-coated plates. Hemagglutinin-neuraminidase (HN) of the virus serves as a ligand structure for NKp44 and NKp46, as indicated by the blockade of binding to NDV-infected cells and viral particles in the presence of anti-HN antibodies and by binding to cells transfected with HN cDNA. Consistent with the recognition of sialic acid moieties by the viral lectin HN, the binding of NKp44-Fc and NKp46-Fc was lost after desialylation. NKp44- and NKp46-CD3zeta lacZ-inducible reporter cells were activated by NDV-infected cells. NDV-infected tumor cells stimulated NK cells to produce increased amounts of the effector lymphokines gamma interferon and tumor necrosis factor alpha. Primary NK cells and the NK line NK-92 lysed NDV-infected tumor cells with enhanced efficiency, an effect that was eliminated by the treatment of target cells with the neuraminidase inhibitor Neu5Ac2en. These results suggest that direct activation of NK cells contributes to the antitumor effects of NDV.


Assuntos
Infecções por Avulavirus/imunologia , Proteína HN/imunologia , Células Matadoras Naturais/imunologia , Vírus da Doença de Newcastle/enzimologia , Infecções por Avulavirus/virologia , Linhagem Celular Tumoral , Proteína HN/genética , Humanos , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Receptor 1 Desencadeador da Citotoxicidade Natural/imunologia , Receptor 2 Desencadeador da Citotoxicidade Natural/genética , Receptor 2 Desencadeador da Citotoxicidade Natural/imunologia , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia
13.
Int J Oncol ; 33(4): 823-32, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18813797

RESUMO

A new recombinant (rec) Newcastle disease virus (NDV) with incorporated human interleukin 2 (IL-2) as foreign therapeutic gene [rec(IL-2)] will be described. The foreign gene in rec(IL-2) did not affect the main features of NDV replication nor its tumor selectivity. Biologically active IL-2 was produced in high amounts by tumor cells infected with rec(IL-2). Tumor vaccine cells infected by rec(IL-2) stimulated human T cells to exert anti-tumor activity in vitro in a tumor neutralization assay. These effects were significantly increased when compared to vaccine infected by rec(-) virus without IL-2 gene. After incubation with rec(IL-2) infected tumor cells, T cells showed increased expression of the activation marker CD69 and produced increased amounts of IFNgamma when compared to T cells co-incubated with rec(-) infected tumor cells. CD8 T cells incubated with rec(IL-2) infected tumor cells showed upregulation of perforin, cell surface exposure of the degranulation marker CD107a and increased anti-tumor cytotoxic activity. Purified T cells from lymph nodes of head and neck squamous cell carcinoma (HNSCC) patients could be stimulated to secrete IFNgamma in an ELISPOT assay upon 40 h of stimulation with rec(IL-2) infected autologous tumor cells [ATV-rec(IL-2)] but not upon stimulation with rec(IL-2) infected allogeneic U937 tumor cells. This suggests direct activation of patient derived tumor antigen-specific memory T cells by ATV-rec(IL-2). In conclusion, the already inherent immunostimulatory properties of NDV could be further augmented by the introduction of the therapeutic gene IL-2. Active specific immunization of patients with ATV-rec(IL-2) should provide the microenvironment at the vaccination site with IL-2 and avoid side effects as seen after systemic IL-2 application.


Assuntos
Vacinas Anticâncer/metabolismo , Interleucina-2/metabolismo , Vírus da Doença de Newcastle/genética , Linfócitos T/virologia , Antígenos CD/biossíntese , Antígenos de Diferenciação de Linfócitos T/biossíntese , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/virologia , Linhagem Celular Tumoral , Citometria de Fluxo , Neoplasias de Cabeça e Pescoço/imunologia , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Lectinas Tipo C , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Ativação Linfocitária , Ligação Proteica , Linfócitos T/metabolismo , Células U937
14.
Int J Cancer ; 119(2): 328-38, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16470838

RESUMO

To investigate tumor-selective viral replication, we compared several tumorigenic human cell lines to nontumorigenic human cells from the blood for the sensitivity to become infected by a recombinant lentogenic strain of Newcastle Disease Virus (NDV) with incorporated transgene EGFP (NDFL-EGFP). Although fluorescence signals in nontumorigenic cells were only weak or missing completely, a massive and long-lasting transgene-expression was observed in all tumor cell lines. The majority of tumor cells (50-95%) could be infected, and viral replication was associated with an increase in the cell surface density of viral antigens. To clarify the underlying mechanism of the observed difference in virus susceptibility we examined the kinetics of interferon-induced antiviral enzymes because NDV is a strong type-I interferon inducer. This analysis revealed several defects of tumor cells in their antiviral defence responses: They showed no response to UV-inactivated NDV, whereas nontumorigenic cells reacted with induction of high-levels of the antiviral enzymes PKR and MxA. Upon coincubation with live NDV, tumor cells showed a delayed response in the increased expression of the antiviral enzymes in comparison with PBMC. In nontumorigenic cells the replication cycle of NDV stopped after the production of positive-strand RNA, while tumor cells continued in the replication cycle and copied viral genomes 10-50 hr after infection. These results can explain the tumor selective replication behavior of this interesting antineoplastic virus.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Proteínas de Fluorescência Verde/genética , Vírus da Doença de Newcastle , Transgenes , eIF-2 Quinase/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon alfa-2 , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Proteínas de Resistência a Myxovirus , Vírus da Doença de Newcastle/genética , RNA Viral , Proteínas Recombinantes , Fatores de Tempo , Regulação para Cima , eIF-2 Quinase/efeitos dos fármacos
15.
Int J Cancer ; 118(3): 658-67, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16108015

RESUMO

We recently reported on newly designed virus-targeted bispecific CD3- and CD28-binding molecules for human T-cell activation. When bound via one arm to a human virus-modified tumor cell vaccine, these reagents caused a polyclonal T-cell response and overcame the potential various T-cell evasion mechanisms of tumor cells. In our current study, we demonstrated the induction of strong antitumor activity in human lymphocytes upon coincubation with a virus-modified tumor vaccine containing anti-CD3 and anti-CD28 bispecific antibodies. Blood mononuclear cells or purified T cells that were coincubated with such a tumor vaccine for 3 days were able to destroy monolayers of human breast carcinoma and other carcinoma cells. Serial transfer to new tumor cell monolayers revealed antitumor cytotoxic activity in such effector cells that lasted for about 10 days. Nontumor target cells appeared to be much less sensitive to the activated effector cells. Although the bispecific molecules alone did not activate effector cells, their binding to virus-infected tumor cells was important and more effective than their binding to free virus. Antitumor activity of the activated effector cells was mediated through soluble factors as well as through direct cell contact of effector cells with the nontargeted bystander tumor cells. Since the virus-modified tumor vaccine is well tolerated and already exhibits a certain effectiveness in cancer patients, the combination with new bispecific molecules has the potential to introduce additional antitumor effects. The reagents can also be combined with Newcastle Disease Virus (NDV)-based oncolytic virotherapy.


Assuntos
Anticorpos Biespecíficos/imunologia , Neoplasias da Mama/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Vacinas Anticâncer , Linfócitos T/imunologia , Neoplasias da Mama/terapia , Proliferação de Células , Feminino , Humanos , Hibridomas , Imunofenotipagem , Imunoterapia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Células Tumorais Cultivadas
16.
Vaccine ; 23(19): 2439-53, 2005 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-15752830

RESUMO

The aim was to develop T cell costimulatory molecules that are broadly applicable to augment anti-tumor immune responses upon application of a virus-modified tumor vaccine to cancer patients. We generated recombinant bispecific single-chain antibodies with one specificity directed against the CD3 or the CD28 antigen on human T cells and the other against the viral target molecule hemagglutinin-neuraminidase (HN) of Newcastle Disease Virus (NDV). By re-directing unstimulated primary human T cells against HN-expressing NDV-infected tumor cells, the bispecific molecule bsHN-CD3 cross-linked effector and target cells and rapidly induced cytotoxicity at nanomolar concentrations. The bsHN-CD28 molecule exerted T cell co-stimulatory function. Maximal T cell activation was achieved with tumor cells infected by NDV and modified with both new stimulatory molecules. This was revealed by T cell proliferation, upregulation of CD69 and CD25 and by release of cytokines, interferons and chemokines. The new molecules combine high-effectivity with specificity and safety.


Assuntos
Anticorpos/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Vacinas Anticâncer/imunologia , Vírus da Doença de Newcastle/imunologia , Linfócitos T/imunologia , Animais , Anticorpos/genética , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Antígenos Virais/genética , Antígenos Virais/imunologia , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Citocinas/análise , Citotoxicidade Imunológica , Proteína HN/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Lectinas Tipo C , Ativação Linfocitária , Linfocinas/análise , Camundongos , Receptores de Interleucina-2/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...